Quilts for pancreatic cancer patients and research – Royal Society of Chemistry

Dr Clare Hoskins, a Reader in Strathclydes Department of Pure and Applied Chemistry, is leading Quilts4Cancer, along with Dr Kirsty Ross, the Departments Outreach Officer. The programme is being launched to coincide with Pancreatic Cancer Awareness Month in November.

Dr Hoskins said: "Funds for cancer research have dropped significantly in the pandemic but we aim to raise awareness of chemistry research across the UK towards better therapies for pancreatic cancer, as well as raising awareness of its signs and symptoms.

"We chose to have quilts made because its known that chemotherapy patients get cold during their treatment. The therapy has serious side effects but we want to raise awareness of the new technology and compounds coming through; a lot of work is being done on making treatment safer, with fewer side effects.

"Were asking for researchers in the chemical sciences to send simple diagrams based on their research, with small summaries of what it is and who is carrying it out. It could show the chemical structures of treatments or technology such as ultrasound equipment.

Picture: University of Strathclyde

Here is the original post:
Quilts for pancreatic cancer patients and research - Royal Society of Chemistry

Global Healthcare Nanotechnology (Nanomedicine) Market Scope 2021-2027 Industry Growth, Business Opportunity, and Leading Players Industrial IT -…

Research Report on Healthcare Nanotechnology (Nanomedicine) Market added by In4Research consist of Growth Opportunities, Development Trends, and Forecast 2026. The Global Healthcare Nanotechnology (Nanomedicine) Market report covers a brief overview of the segments and sub-segmentations including the product types, applications, companies & regions. This report describes the overall Healthcare Nanotechnology (Nanomedicine) Market size by analyzing historical data and future forecast.

The Healthcare Nanotechnology (Nanomedicine) Market Report includes:

To Get Sample Copy of Healthcare Nanotechnology (Nanomedicine) Market Report with Complete TOC, Figures & Graphs Connect with us at https://www.in4research.com/sample-request/23369

Major Key Players Covered in The Healthcare Nanotechnology (Nanomedicine) Market Report include

Healthcare Nanotechnology (Nanomedicine) Market Segments and Sub-segments Covered in the Report are as per below:

By Type:

By Application:

For more Customization, Connect with us at https://www.in4research.com/customization/23369

Geographically, this report is segmented into several key Regions along with their respective countries, with production, consumption, revenue (million USD), and market share and growth rate of Healthcare Nanotechnology (Nanomedicine) in the following regions:

The Covid19 pandemic has transformed the market landscape. The market ecosystem has taken a directional shift in the way supply-side of the market is accessed. The report covers the aftermath of the Covid19 catastrophe.

Get the PDF to understand the CORONA Virus/COVID19 impact and be smart in redefining business strategies: https://www.in4research.com/impactC19-request/23369

Important Features that are under offering & key highlights of the Healthcare Nanotechnology (Nanomedicine) Market report:

Any Questions/Queries or need help? Speak with our analyst https://www.in4research.com/speak-to-analyst/23369

Major Points in Table of Content of Healthcare Nanotechnology (Nanomedicine) Market

Chapter 1. Research Objective

1.1 Objective, Definition & Scope

1.2 Methodology

1.3 Insights and Growth Relevancy Mapping

1.4 Data mining & efficiency

Chapter 2. Executive Summary

Chapter 3. Strategic Analysis

3.1 Healthcare Nanotechnology (Nanomedicine) Market Revenue Opportunities

3.2 Cost Optimization

3.3 Covid19 aftermath Analyst view

3.4 Healthcare Nanotechnology (Nanomedicine) Market Digital Transformation

Chapter 4. Market Dynamics

4.1 DROC

Chapter 5. Segmentation & Statistics

5.1 Segmentation Overview

5.2 Demand Forecast & Market Sizing

Chapter 6. Market Use case studies

Chapter 7. KOL Recommendations

Chapter 8. Investment Landscape

8.1 Healthcare Nanotechnology (Nanomedicine) Market Investment Analysis

8.2 Market M&A

8.3 Market Fund Raise & Other activity

Chapter 9. Healthcare Nanotechnology (Nanomedicine) Market Competitive Intelligence

9.1 Company Positioning Analysis

9.2 Competitive Strategy Analysis

Chapter 10. Company Profiles

Chapter 11. Appendix

Buy Full Report at: https://www.in4research.com/buy-now/23369

Key Benefits of the Report:

For More Details Contact Us:

Contact Name: Rohan

Email: [emailprotected]

Phone: +1 (407) 768-2028

Go here to read the rest:
Global Healthcare Nanotechnology (Nanomedicine) Market Scope 2021-2027 Industry Growth, Business Opportunity, and Leading Players Industrial IT -...

Combination Therapy of Carfilzomib and Paclitaxel for PACs | IJN – Dove Medical Press

Introduction

Pancreatic adenocarcinomas (PACs) are relatively rare compared to other cancers and represent only 3.2% of all new cancer cases in the US. Nevertheless, the average 5-year survival rate for all stages of PAC is only 10.0% because PACs cannot be detected and treated early. In general, the percent of cases and 5-year relative survival according to various stages of PAC (localized, regional, distant, and unknown) at diagnosis were reported to be 11% and 39.4%, 30% and 13.3%, 52% and 2.9%, and 7% and 6.1%, respectively, showing that the earlier PAC is caught, the better chance a person has of surviving 5 years after being diagnosed.1 Gemcitabine (GEM) is the only approved first-line monotherapy for treating PACs. Unfortunately, it still delivers unsatisfactory therapeutic outcomes in prolonging progression-free survival (PFS) and overall survival (OS) of patients with locally advanced and metastatic PAC.2

Combined therapy has become a major means to combat cancer thanks to its primary advantages of increased efficacy without, or with minimal, addictive toxicities at equal or reduced administered doses. Multiple combination therapies composed of GEM and different cytotoxic and biologic agents have undergone clinical evaluations to examine the therapeutic efficacies for patients at various stages of PAC since 2002 as reported by Lei et al.3 Among them, only the combination regimen of GEM with either erlotinib (Tarceva) or paclitaxel albumin-bound nanoparticles (NPs) (Abraxane) demonstrated significant improvements in most clinical outcome parameters compared to GEM alone, leading to the approval of both combination therapies by the US Food and Drug Administration (FDA) as a first-line combination therapy for patients with locally advanced and metastatic PAC in 20154 and 2013,5 respectively. Despite combination therapies having demonstrated improved outcomes in patient survival and quality of life, the overall improvement is still marginal, especially for patients diagnosed with late stages of the disease. There is still an urgent need to generate effective strategies, new single agents, or new combinations, to significantly improve clinical outcomes for treating PACs.

Several studies support the proteasome being an effective therapeutic target against PAC from the perspectives of high heterogeneity and chemoresistance. By unraveling transcriptomic predictive signature data by Fraunhoffer et al,6 a subgroup of PACs sensitive to FDA-approved carfilzomib (CFZ) was identified, and it was ultimately suggested to repurpose CFZ for treating PACs.7 Furthermore, proteasome inhibitors (PIs), such as carfilzomib, can induce apoptosis in PACs by inducing endoplasmic reticular (ER) stress, which facilitates synergistic effects when combined with radiation therapy or chemodrugs like camptothecin and paclitaxel.8 It was further disclosed that a combination of MG-132 (a PI) and camptothecin at a ratio of 5:1 (2.5 mol/l MG-132: 0.5 mol/l camptothecin) provided promising results with enhanced cytotoxicity compared to the single compounds in MIA PaCa-2 cells, while that for the combination of MG-132 and paclitaxel at the same 5:1 ratio but with lower concentrations of 0.08 mol/l MG-132 and 0.016 mol/l paclitaxel could moderately increase the cytotoxicity to 62% from 46% for paclitaxel alone at the same concentration of 0.016 mol/l as that in combination.9 This potentially suggests that a combination of PIs, such as CFZ with paclitaxel for treating PACs might be worth pursuing.

The poor biostability and short half-life of CFZ are considered major issues causing CFZ to perform with low efficacy in patients with solid cancers because it is difficult for CFZ to arrive at the proteasome in solid tumors.10,11 Polymer micelles (PMs) composed of biodegradable block copolymers poly(ethylene glycol) (PEG) and poly(caprolactone) (PCL) were reported to improve the metabolic stability of CFZ in vitro. However, despite the in vitro metabolic protection of CFZ, CFZ-loaded PMs or PEG-PCL-deoxycholic acid (CFZ-PMs) did not display superior in vivo anticancer efficacy in mice bearing human lung cancer xenografts (H460) to that of the clinically used cyclodextrin-based CFZ (CFZ-CD) formulation.12 A novel albumin-coated nanocrystal formulation of CFZ (CFZ-alb NC) displayed improved metabolic stability and enhanced cellular interactions, uptake, and cytotoxic effects in breast cancer cells in vitro. Consistently, CFZ-alb NCs showed greater anticancer efficacy in a murine 4T1 orthotopic breast cancer model than the currently used cyclodextrin-based formulation. It was highly suggested that human serum albumin (HSA)-bound NPs could be used as a viable nanocarrier to encapsulate CFZ for cancer therapy.13 As described above, it was paclitaxel (PTX) albumin-bound nanoparticles (Abraxane), not Taxol (a traditional dosage form with PTX being dissolved in the mixture of Cremophor EL and ethanol), which was approved for combination with GEM by the FDA as a first-line combination therapy for patients with locally advanced and metastatic PACs. Therefore, HSA-bound NPs could be used as a viable nanocarrier to co-encapsulate PTX and CFZ for combination therapy of PAC.

HSA has garnered considerable interest as a nanocarrier, due to its low toxicity, biocompatibility, and the ability to reduce interactions with phagocytes in the reticuloendothelial system (RES).1416 Moreover, albumin can interact with cancer cells based on its increased use as an energy source in rapidly proliferating cancer cells.17 It was reported that nanoalbumin-bound (nab)-drugs can aid drug permeation across tumor vessels.18,19 It was also suggested that albumin facilitates the movement of nab-drugs across endothelial cell membranes by binding to the gp60 receptor and sequentially interacting with other albumin-binding proteins such as secreted protein acidic and rich in cysteine (SPARC), which is abundantly expressed in and near cancer cells.2022 As exemplified, GEM (Gemcitabine)-loaded HSA and PTX-loaded HSA for practical PAC treatment have been reported by Han et al and Yu et al, respectively.23,24 Therefore, it was thought that NPs fabricated with HSA might potentially be an optimal choice for co-delivery of chemotherapeutic drugs with a high drug loading capacity, biodegradability, and good biocompatibility.

Abraxane is the first FDA-approved chemotherapeutic formulation based on Nab nanotechnology, which relies heavily on the use of organic solvents, namely, chloroform.25 The toxicity introduced by residual chloroform poses a potential risk to patient health. In response to the issue of chronic toxicity, a reversible self-assembling method, which eliminates the dependence on toxic organic solvents during manufacturing, was developed in a preliminary study and demonstrated to be capable of successfully preparing HSA-bound PTX and CFZ nanosuspensions. Furthermore, both NPs formed using this method still retained their suitability for intravenous (IV) administration.25 Therefore, in this study, the preparation of CFZ-loaded, PTX-loaded, and CFZ/PTX co-loaded HSA NPs was developed and optimized. To confirm these advantages, the properties of the three drug-loaded HSA NPs, including the encapsulating efficiency (EE), drug-loading (DL), mean size, polydispersity index (PDI), drug release, and cell growth inhibition against MIA CaPa-2 cells (human pancreatic cancer cell line) were characterized in vitro. Furthermore, the in vivo pharmacokinetic study of the three drug-loaded HSA NPs (CFZ/HSA NPs, PTX/HSA NPs, and CFZ/PTX/HSA NPs) were evaluated in Sprague-Dawley rats and compared to two solvent-based (Sb) drugs of CFZ and PTX (Sb-CFZ and Sb-PTX). The anti-tumor efficacy and systemic toxicity were further evaluated in MIA CaPa-2 tumor-bearing C.B-17 SCID mice.

Sb-CFZ (solvent-based CFZ) was prepared by dissolving 60 mg of CFZ (Chunghwa Chemical Synthesis & Biotech, New Taipei City, Taiwan), 3000 mg sulfobutylether beta-cyclodextrin (SBE--CD), and 57.7 mg citric acid in 29 mL deionized water through sodium hydroxide (NaOH) pH adjustment (pH=3.5). The solution was lyophilized and stored at 4C until reconstitution for use.26 Sb-PTX (solvent-based PTX) was prepared by solubilizing 6 mg PTX (ScinoPharm, Tainan, Taiwan) in 527 mg of purified Cremophor EL (polyoxyethylated castor oil; BASF, Ludwigshafen, Germany) in 497 mg (v/v) of dehydrated alcohol.

Drug-loaded HSA NPs were prepared with defatted human serum albumin (HSA) by a self-assembling method developed in our lab. Defatted HSA was produced by adsorption of fatty acids in HSA onto charcoal as previously described.27 Briefly, a marketed 20% HSA solution (Taiwan Blood Services Foundation, Taipei, Taiwan) was diluted with deionized water, and then the pH was adjusted to 2.7 with 1 N HCl. After adding 5 g of activated charcoal, the resulting HSA solution was stirred at 300 rpm and 4C for 2 h. The mixed solution was centrifuged at 8000 rpm and 4C for 10 min, and the supernatant was filtered through a 0.45-m nylon membrane (ChromTech, Bad Camberg, Germany) to remove the charcoal. Finally, the pH of the filtrate was adjusted to 7.0 with 1 N NaOH and lyophilized. The so-obtained lyophilized HSA powder was stored at 4C.28

The preparation of drug-loaded HSA NPs was divided into three steps. First, the pH of the HSA solution was adjusted to 2.7 with 1.0 N HCl to expose the hydrophobic domains.27,28 Second, the targeted hydrophobic drug in ethanol was added followed by stirring for 5 min to enhance interactions between the drug and HSA. Finally, the pH value was re-adjusted to neutral with 0.1 N NaOH to induce self-assembling and encapsulate the hydrophobic drug. Then, used high-pressure homogenization with an N2-3D Nanolyzer (Gogene, Hsinchu, Taiwan) to form stabilized drug-loaded HSA NPs. An Amicon Ultra-15 centrifugal filter (with a molecular weight (MW) cutoff of 10 kDa) was used to remove the ethanol, salt, and free drug, and then the drug-loaded HSA NPs were concentrated. The drug-loaded HSA NP concentrate was passed through 0.20-m regenerated cellulose filtration (Phenomenex, Torrance, CA, USA) to obtain translucent dispersion with typical diameter around 150 nm. Finally, lyophilized the solution for 48 hours without cryoprotectant.

The formulation and optimal homogenizer parameters utilized in step 3 for preparing CFZ-loaded HSA NPs (CFZ/HSA NP), PTX-loaded HSA NPs (PTX/HSA NP), and CFZ/PTX-loaded HSA NPs (CFZ/PTX/HSA NP) are described below. To prepare CFZ/HSA NPs, 200 mg CFZ was dissolved in 20 mL absolute alcohol, and the dispersion was added to 200 mL 0.9% defatted HSA solution. The ratio of drug to HSA was 1:9, and a 10K psi homogenizer parameter was applied for 10 cycles. To prepare PTX/HSA NPs, 300 mg PTX was dissolved in 12 mL absolute alcohol, and the dispersion was added to 120 mL 2.25% defatted HSA solution. The ratio of drug to HSA was also 1:9, and the 20K psi homogenizer parameter was applied for 20 cycles. To prepare CFZ/PTX/HSA NPs, 60 mg CFZ and 120 mg PTX were dissolved in 18 mL absolute alcohol, and the dispersion was added to 180 mL 1% defatted HSA solution. The ratio of both drugs to HSA was 1:10, and the 10K psi homogenizer parameter was applied for 10 cycles.

The mean particle size, size distribution, zeta potential, and polydispersity index (PDI) of drug-loaded HSA NPs were measured with a Zetasizer nano ZS (Malvern, Worcestershire, UK) by scattering angle of 90 at 25C. The drug-loaded HSA NPs were diluted with double-distilled water before the measurement, and all measurements were performed at least in triplicate. The shape and size were also observed by transmission electron microscopy (TEM), using Hitachi H-7000 (Hitachi, Tokyo, Japan). The purified NPs were diluted with water to allow clearer pictures to be taken. Samples were prepared by placing a drop on carbon-coated copper grids and sponging off the excess with filter paper. Then, the samples were stained with uranyl acetate (2% aqueous solution) for 3 minutes and dried at room temperature.28

To assess the entrapment efficiency of CFZ or PTX in drug-loaded HSA NPs, 10 mg lyophilized NPs was dissolved in 1 mL deionized water; then 9 mL acetonitrile was added and vortexed it for 1 minute. The solution was centrifuged at 14,000 rpm for 10 minutes. After appropriate dilution, CFZ or PTX in the supernatant was directly quantified by Waters alliance HPLC (Waters, Milford MA, USA) equipped with an Inert Sustain C18 column (150 4.6 mm, particle size 5 m, GL Sciences, Tokyo, Japan). The mobile phase was composed of acetonitrile and 0.05% formic acid aqueous solution (50:50, v/v, at a flow rate of 1 mL/min). The total analytical time for a single injection was 12 min. The injection volume was 10 L, and chose 210-nm wavelength for detection. The column oven was kept at 35C, and the sample cooler was maintained at 10C. The drug loading (DL) and entrapment efficiency (EE) of nanoparticles were calculated by the following equations:

and

;

WM is the weight of the drugs in the NPs, WI is the weight of the initial feeding drug, and WP is the weight of the initial feeding HSA.

Cell viabilities of the CFZfree (CFZ dissolved in DMSO), Sb-CFZ, CFZ/HSA NPs, PTXfree (PTX dissolved in DMSO), Sb-PTX, PTX/HSA NPs, and CFZ/PTX/HSA NPs were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay for MIA PaCa-2 cell line obtained from ATCC. Cells were seeded at a density of 3 103 cells/well in 96-well plates and incubated for 24 h at 37C with 5% CO2. Then, tumor cells were treated with different concentrations (0.01, 0.1, 1, 5, 10, 50, 100, and 1000 ng/mL) of CFZfree, Sb-CFZ, PTXfree, Sb-PTX, or drug-loaded HSA NPs. After incubation for 72 hours, 200 L MTT (0.5 mg/mL) was added to each well for 2 hours. After removing the medium, 50 L DMSO was added to each well and gently shaken to dissolve any purple formazan crystal. The absorbance was measured at 550 nm (Bio-Tek). The survival rate was calculated using the following formula: percentage (%) cell survival = [(mean absorbency in test wells)/(mean absorbency in control wells)] 100. Values of the combination index (CI) were calculated by the Chou-Talalay method:29,30

C and P denote IC50 values of CFZ and PTX in combination therapy that inhibits 50% of the cell. C50 and P50 denote doses of CFZ and PTX that inhibit 50% cells alone. Values of CI = 1, CI < 1, and CI > 1, respectively, indicate additivity, synergy and antagonism.

Drugs released from the formulations were investigated in PBS (containing 0.5% Tween 80) by the dialysis method. The CFZ- or PTX-loaded HSA NPs were diluted to 0.1 or 0.2 mg/mL in 1-mL solution and then placed in a dialysis bag (OrDial D80-MWCO 60008000, cat. no. 60082530, Orange Scientific, Braine-lAlleud, Belgium) against 40 mL release medium, with 100 rpm shaking speed at 37C. Sampled 1 mL at 1, 2, 3, 4, 6, 8, 12, 24, and 48 hours, and performed the analysis using the HPLC method above.31

Male BALB/c mice (BioLasco Taiwan, Yilan, Taiwan) at 7 weeks of age were randomized into 4 groups and each group contained 4 mice. For single-dose study, we used tail vein injection at 0 days. For multi-dose study, we repeated half dose of single-dose study at 0 and 1 day. If there was no obvious toxic reaction, the dosage was elevated correspondingly. The weight changes and physiological signs were observed and recorded for 5 consecutive days in the first week. During the second week, the related assessment would be performed every 2 days. The whole study continued for 15 days. It would be specified as the maximum tolerance dose if there is any event for neurotoxicity, weight loss >20% or death.

Male Sprague-Dawley rats (BioLasco Taiwan, Yilan, Taiwan) at 8~10 weeks of age were used to study pharmacokinetic profiles after administration of Sb-CFZ, Sb-PTX, CFZ/HSA NPs, PTX/HSA NPs, Sb-CFZ+Sb-PTX, CFZ/HSA NPs+PTX/HSA NPs, and CFZ/PTX/HSA NPs. Rats were given a single tail vein injection of 5 mg/kg CFZ and 10 mg/kg PTX for each formulation (three or four rats per group). Blood samples were collected from the jugular vein in heparinized tubes at 0.017, 0.033, 0.083, 0.25, 0.5, 1, 2, 4, 6, 8, 12, 24, and 48 h after administration. All blood samples were immediately centrifuged at 4500 rpm for 10 minutes to obtain plasma, and then stored at 80C until analyzed by UPLC interfaced with MS (Triple Quadrupole Mass Spectrometry, TQ-XS, Waters). CFZ and PTX were extracted from the plasma as follows: 100L plasma was extracted with tert-butyl methyl ether (400 L) containing an internal standard (500 ng/mL chlorpropamide (Sigma-Aldrich, St. Louis, MO, USA) and 500 ng/mL docetaxel (ScinoPharm Taiwan, Tainan, Taiwan)) by vortex-mixing for 1 min. After centrifugation at 14,000 rpm for 10 min, 300 L organic phase was transferred to a new tube and dried at 40C. Samples were reconstituted in 100 L mobile phase and transferred to a new vial for the UPLC-MS/MS analysis. The measurement by UPLC-MS/MS. Chromatographic separation was performed with a Purospher Star RP-18 end-capped column (2.1 50 mm, particle size 2 m, Merck) and gradient elution (at a flow rate of 0.3 mL/min). The mobile phase comprised 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B). The total analytical time for a single injection was 5 minutes, and the injection volume was 2 L. The column oven was kept at 40C, and the sample cooler was kept at 10C. Detection of ions was performed in the positive ionization mode with the following transitions in multiple reaction monitoring mode (MRM): 720.33 100.03 for CFZ, 277.06 174.87 for chlopropamide (the internal standard for CFZ),32 854.29 104.99 for PTX, and 830.40 549.24 for docetaxel (the internal standard for PTX). The capillary voltage was 3.0kV, cone voltage was 30V, desolvation temperature was 350C, desolvation gas flow was 650L/h, and collision gas flow was 25L/h.

In vivo pharmacokinetic parameters including the area under the plasma concentrationtime curve (AUC), the apparent volume of distribution (V), plasma clearance (CL), and elimination half-life (T1/2) of each formulation were calculated and expressed by the mean and standard deviation (SD). The AUC0-1h, AUC0-2h, AUC0-24h, and AUC0-infinity were estimated by linear trapezoidal method. Plasma clearance (CL) was calculated from the dose/AUC0-infinity. The initial half-life (T1/2,initial) and terminal half-life (T1/2,terminal) values were calculated as ln(2)/k, where k represents either the initial distribution rate constant or the terminal elimination rate constant obtained from the slope of a semilogarithmic plot of the concentrationtime profile. The volume distribution (V) was estimated using a noncompartmental method provided by WinNonlin software (vers. 6.3.0.395, Pharsight, Princeton, NJ, USA). The maximum plasma concentration (Cmax) was recorded as observed for the first sampling time point, and C0 was the concentration at t = 0 (extrapolated).

C.B-17 female SCID mice, at 6~7 weeks of age (BioLasco Taiwan), were used as the tumor xenograft models. The models were established by subcutaneously inoculating MIA PaCa-2 (2107 cells/mouse, 100-L injection) into the right dorsal flank of each mouse. MIA PaCa-2 tumor-bearing mice with 150 mm3 tumor volumes were randomly divided into eight treatment groups (n = 5). One group of mice received an intravenous injection of saline as a control. The other groups received an injection of Sb-CFZ, Sb-PTX, CFZ/HSA NPs, PTX/HSA NPs, Sb-CFZ+Sb-PTX, CFZ/HSA NPs+PTX/HSA NPs, or CFZ/PTX/HSA NPs (equivalent to 5 mg/kg CFZ and 10 mg/kg PTX for each mouse). Administration was performed on days 0, 1, 7, 8, 14, and 15. Body weights (BWs) and tumor sizes were measured three times every week using digital calipers, and tumor volumes (mm3) were calculated. After being sacrificed by CO2 on day 46, tumors were harvested and weighed. The tumor growth inhibition (TGI) (%) was calculated as follows:33

Female C.B-17 SCID mice, at 6~7 weeks of age (BioLasco Taiwan), were used as the tumor xenograft models. The models were established by subcutaneously inoculating MIA PaCa-2 cells (2107 cells/mouse, 100-L injection) in the right dorsal flank of each mouse. On day 14 after tumor cell inoculation when tumor volumes had reached about 150 mm3, each mouse was given Sb-CFZ, Sb-PTX, CFZ/HSA NPs, PTX/HSA NPs, Sb-CFZ+Sb-PTX, CFZ/HSA NPs+PTX/HSA NPs, or CFZ/PTX/HSA NPs (equivalent to 5 mg/kg CFZ and 10 mg/kg PTX in each mouse) by an intravenous injection. After 2 and 8 h, mice were sacrificed by anesthesia and perfused with a PBS solution to remove the blood. The heart, lungs, liver, spleen, kidneys, and tumors were excised, weighed, and stored at 80C. Tissues were homogenized by an ultrasonicator probe (VCX 750; Sonics & Materials, Newtown, CT, USA) with 5 W and three pulses for 10 s. After that, 400 L of a PBS/0.1% heparin solution was added. Tissue homogenates (200 L) were obtained, and drug concentrations were analyzed by UPLC/MS/MS.

Data are presented as the mean SD of three different replicates. For in vivo studies, a one-way analysis of variance (ANOVA) with Tukeys multiple comparisons was used to test for significant differences in the longitudinal tumor volume growth over the entire experimental period among the eight treatment groups and to determine whether there was a significant interaction effect between CFZ and PTX. Significant differences between groups were indicated by *p<0.05 and **p<0.005.

As depicted in Figure 1, the mean particle sizes of CFZ/HSA NPs, PTX/HSA NPs, and CFZ/PTX/HSA NPs were 114.50.6, 117.40.4, and 105.30.6 nm; PDI values were 0.1440.007, 0.1660.007, and 0.1670.008; zeta potentials were 23.000.70, 21.10.58, and 21.30.70 mV; entrapment efficiencies (EEs, %) were 95.62.1%, 97.13.5%, and 92.72.6%/90.73.1%; and drug loadings (DLs) were 9.40.1%, 10.10.2%, and 9.10.4%, respectively. In addition, all zeta potentials measured were between 21.1 and 23.00 mV and indicated that the drug was encapsulated with HSA, which possessed a negative charge at a neutral pH because its PI was equivalent to 4.7.34 Figure 1 demonstrates that the three different HSA NPs were spherical, and the mean particle sizes were <200 nm under 80,000 TEM observation. Another noteworthy result was that the entrapment efficiencies for CFZ and PTX were, respectively, observed to be 92.72.6% and 90.73.1% in CFZ/PTX/HSA NPs. The calculated ratio for PTX and CFZ was 1.96; in other words, the method was suitable for preparing HSA NPs by achieving designed ratio nearly 2. Also, the high entrapment efficiency and drug loading (9.1%) indicated that HSA could act as novel and excellent nanocarriers for co-loading two drugs. In addition to the good compatibility with the two hydrophobic drugs, the preparation is simple and efficient with no cryoprotectant required.

Figure 1 TEM image and particle size analysis of drug/human serum albumin (HSA) nanoparticles (NPs).

The MIA PaCa-2 cell line was used as a model in the cytotoxicity study. Table 1 shows the values of the 50% inhibitory concentration (IC50) for different combinations of free drug in DMSO (CFZfree and PTXfree), two solvent-based drugs (Sb-CFZ and Sb-PTX), and two drug-loaded HSA NPs (CFZ/HSA NPs and PTX/HSA NPs). Respective IC50 values for CFZfree, Sb-CFZ, CFZ/HSA NPs, PTXfree, Sb-PTX, and PTX/HSA NPs were 8.9, 8.12, 7.89, 0.44, 0.47, and 0.86 ng/mL. The results indicated that the two model drugs maintained similar cytotoxicities in the different formulations. Compared to CFZ, PTX demonstrated a higher cytotoxicity toward the MIA PaCa-2 cell line. Table 1 also reveals the synergism of CFZ and PTX at ratios of 1:2, 1:1, and 2:1. IC50 values for free CFZ and PTX ratios of 1:2, 1:1, and 2:1 were 0.25, 1.39, and 1.00 ng/mL, respectively. According to the equation of the Chou-Talalay method, synergisms existed with free CFZ and PTX ratios of 1:2 and 2:1, and CI50 values were 0.38 and 0.83. For the Sb-CFZ and Sb-PTX combination ratios of 1:2, 1:1, and 2:1, IC50 values were 0.03, 0.37, and 0.51 ng/mL, and CI50 values were 0.05, 0.42, and 0.40, respectively. These results also revealed the synergism of Sb-CFZ and Sb-PTX. As for CFZ and PTX HSA NP ratios of 1:2, 1:1, and 2:1, IC50 values were 0.01, 0.07, and 0.66 ng/mL, and CI50 values were 0.01, 0.04, and 0.31, respectively. Figure 2 illustrates that the original product and HSA combinations exhibited synergism in all ratios examined. Since the combination ratio of 1:2 (CFZ: PTX) demonstrated the more-obvious synergic effect with IC50 value of 0.1 ng/mL and CI50 value of 0.08, the co-encapsulated ratio of 1:2 for CFZ and PTX in HSA (CFZ/PTX/HSA NPs) was chosen as the target formulation for the following assessment studies including the drug release study.

Table 1 50% Inhibitory Concentration (IC50) Values of Different Combinations of Carfilzomib and Paclitaxel

Figure 2 Characterization of the synergistic activity of combined carfilzomib (CFZ) and paclitaxel (PTX) treatment at different weight ratios.

Release percentages of CFZ and PTX from various formulations were assessed, and results are shown in Figure 3A and B, respectively. As shown in Figure 3A, the release of CFZ from Sb-CFZ, CFZ/HSA NPs, CFZ/HSA NPs+PTX/HSA NPs, and CFZ/PTX/HSA NPs were observed to have reached a plateau at 12~24 h with similar profiles, and release percentages at 12 h were determined to be 68.6%7.1%, 79.4%4.9%, 71.2%7.9%, and 71.7%3.8%, respectively. Furthermore, over 90% of CFZ had been released from CFZ/HSA NPs and CFZ/HSA NPs+PTX/HSA NPs at 24 h. We observed that the more-rapid release of CFZ from Sb-CFZ might be attributed to the use of the hydrophilic SBE--CD solubilizer to increase the solubility of CFZ in water. Similarly, the more-complete release of CFZ from the two HSA formulations was probably due to both being encapsulated in HSA NPs, which are expected to have greater surface areas for release. However, the release of CFZ from Sb-CFZ+Sb-PTX was measured at only 31.5%7.2% at 12 and 56.3%8.8% at 48 h, which were slower than that of CFZ released from Sb-CFZ at 12 h. This indicates that the addition of Sb-PTX to Sb-CFZ might have retarded the release of CFZ from Sb-CFZ resulting in a smaller release percentage. We suspect that by mixing Sb-CFZ with Sb-PTX, CFZ was encapsulated within the hydrophobic interior of Cremophor micelles, which was used as a solubilizing agent in the Sb-PTX formulation, causing retardation of permeation across the membrane of the dialysis bag to release CFZ.35

Figure 3 Drug release profiles of carfilzomib (CFZ, A) and paclitaxel (PTX, B). *p<0.05 and **p<0.005.

As for the release of PTX revealed by Figure 3B, release percentages of PTX from PTX/HSA NPs, CFZ/HSA NPs+PTX/HSA NPs, and CFZ/PTX/HSA NPs were observed to have reached a plateau at 12 h with similar profiles, and release percentages at the plateau were, respectively, determined to be 73.2%15.3%, 82.2%0.6%, and 65.6%8.8%. However, the release percentages of PTX from Sb-PTX and Sb-CFZ+Sb-PTX followed a gradually increasing trend, but these forms were only able to release 27.3%6.8% and 18.3%6.9%, respectively, at 48 h. Since both Sb-PTX and Sb-CFZ+Sb-PTX contained Cremophor as the solubilizing agent for PTX, it was expected as described above that the release of PTX trapped in Cremophor micelles would be retarded resulting in a slower release rate being observed. Therefore, CFZ and PTX were released more completely from the drug-loaded HSA NPs since both were encapsulated in HSA NPs, which were expected to have greater surface areas for release. Similarly, the greater extents of release percentages of PTX from the three HSA formulations were probably due to all of them being absorbed onto HSA in HSA NPs, which were expected to present as amorphous form to have higher solubility for increasing the extent of release.

In addition, it is worth mentioning that CFZ/PTX ratios released from co-encapsulated HSA NPs (CFZ/PTX/HSA NPs) were about 1.77~2.08 after 8 h of dissolution, which were consistent with the ratio of CFZ/PTX loaded in HSA NPs. However, the ratios of release amounts between CFZ and PTX from Sb-CFZ+Sb-PTX and CFZ/HSA NPs+PTX/HSA NPs were around 1.06~1.18 and 1.63~2.92, respectively, which did not reach the designed optimal ratio of 1:2 for synergism. It was concluded that release from the CFZ/PTX/HSA NP formulation conformed to the design combination ratio of 1:2 for CFZ and PTX to establish a potential synergistic effect.

Maximum tolerance dose for drug-loaded HSA NP and two free drugs as sb-CFZ and sb-PTX was then evaluated in BALB/c mice. Major dose limiting toxicities of CFZ or PTX were determined by neurotoxicity, weight loss >20% or death. According to the result (Table 2), the established maximum tolerance doses for a single dose were: Sb-CFZ 5 mg/kg, CFZ/HSA 17.5 mg/kg, Sb-PTX 20 mg/kg, PTX/HSA 300 mg/kg, Sb-CFZ+Sb-PTX 3.75/7.5 mg/kg, CFZ/HSA+PTX/HSA 10/20 mg/kg and CFZ/PTX/HSA 10/20 mg/kg. For multi-dose were as follows: Sb-CFZ 2.5 mg/kg, CFZ/HSA 5 mg/kg, Sb-PTX 12.5 mg/kg, PTX/HSA 150 mg/kg, Sb-CFZ+Sb-PTX <2.5/5 mg/kg, CFZ/HSA+PTX/HSA 5/10 mg/kg and CFZ/PTX/HSA 5/10 mg/kg. It is demonstrated that the maximum tolerance dose for drug-loaded HSA NP is normally higher than that for solvent-based form of free drug. Compared with solution base form, the combination or co-load HSA NP has at least 2 times higher maximum tolerance dose. Dr. Ernsting reveals that the maximum tolerance dose of single-dose for Abraxane performed in BALB/c is 170 mg/kg. However, in this research, PTX/HSA shows the quite remarkable tolerability from maximum tolerance dose study (300 mg/kg).36 Moreover, Dr. He also found that HSA encapsulation could lower the systemic nervous toxicity from VM-26.37 Taking the advantages with loading multi-drug in HSA NPs, the goal of achieving higher efficacy with lower toxicity was accomplished with such a multi-drug HSA NPs technique platform.

Table 2 Maximum Tolerance Dose Study for Various Combination Ratios of Carfilzomib and Paclitaxel on BALB/c Mice (n = 4)

Drug concentrations in plasma after a single tail vein injection of two solvent-based drugs (Sb-CFZ and Sb-PTX) and three drug-loaded HSA NPs (CFZ/HSA NPs, PTX/HSA NPs, and CFZ/PTX/HSA NPs) with respective dosing amounts of CFZ and PTX equivalent to 5 and 10 mg/kg are demonstrated in Figure 4A for CFZ and Figure 4B for PTX. Calculated pharmacokinetic parameters are listed in Table 3 for CFZ and Table 4 for PTX. As shown in Table 3, there were no significant differences among AUC0-1h, AUC0-24h, and AUC0-infinity obtained for all the various formulations after a single IV bolus administration of CFZ equivalent to 5 mg/kg. This indicates that CFZ was rapidly distributed to tissues and was quickly cleared from the systemic circulation after IV administration, resulting in the most reliable measure of the drugs bioavailability AUC in a period of 0 to 1 h (AUC0-1h) representing nearly the entire extent of the dosing amount of CFZ entering the systemic circulation. It was reported that the in vivo potency of CFZ is determined by the total dose administered (AUC), not Cmax, since CFZ can be rapidly distributed to tissues after IV administration as demonstrated by the potent proteasome inhibition in a variety of tissues.26 Because of this, pharmacokinetic parameters of T1/2,initial (min) and AUC0-1h (hr*g/mL) which are potentially related to the in vivo potency of CFZ were selected for comparison. Results in Table 3 demonstrate that T1/2,initial (min) and AUC0-1h (hg/mL) for CFZ after administration of Sb-CFZ were shorter and lower, respectively, than those for administration of CFZ/HSA NPs (12.722.14 vs 15.191.38 min and 0.2220.034 vs 3.3374.306 hg/mL), while neither of them was much different from administration of Sb-CFZ+Sb-PTX (12.722.14 vs 11.028.98 min and 0.2220.034 vs 0.2410.056 hg/mL). On the other hand, T1/2,initial (min) and AUC0-1h (hg/mL) for CFZ after administration of CFZ/HSA NPs were longer and much higher, respectively, than those with administration of CFZ/HSA NPs+PTX/HSA NPs (15.191.38 vs 9.142.93 min and 3.3374.306 vs 0.0550.009 hg/mL), while they were longer and much higher, respectively, than those with administration of CFZ/PTX/HSA NPs (15.191.38 vs 10.004.08 min and 3.3374.306 vs 0.5370.451 hg/mL).

Table 3 Pharmacokinetic Parameters of Carfilzomib Obtained from a Single Intravenous Bolus Administration of Various Formulations (Equivalent to 5 mg/kg Carfilzomib)

Table 4 Pharmacokinetic Parameters of Paclitaxel Obtained from a Single Intravenous Bolus Administration of Various Formulations (Equivalent to 10 mg/kg Paclitaxel)

Figure 4 Plasma concentrationtime curves of carfilzomib (CFZ, A) and paclitaxel (PTX, B) after intravenous administration at respective doses of 5 and 10 mg/kg to rats.

As previous research reported, almost no CFZ was detected in plasma 30 min after administration with an initial half-life (T1/2,initial) of <20 min.38 By utilizing the same solvent system composed of SBE--CD to solubilize CFZ (Sb-CFZ), a similar T1/2,initial was observed in this study, thereby confirming the suitability of the pharmacokinetic study conducted in this research. As such, a slower terminal elimination rate (T1/2,initial) observed for administration of CFZ/HSA NPs compared to that for administration of Sb-CFZ indicates that encapsulation of CFZ with HSA somewhat protected CFZ from elimination in plasma leading to a longer T1/2,initial. With a longer T1/2,initial, it was expected to have a higher AUC0-1h as Table 3 demonstrates.

Compared to Sb-CFZ, the combined administration of the two solvent-based formulations (Sb-CFZ+Sb-PTX) resulted in a similar AUC0-1h for CFZ but with a slightly lower T1/2,initial for the CFZ distribution into tissue compartments. This might indicate that drugdrug interactions exist between CFZ and PTX that are dissolved in solvent as free solubilized forms leading to an influence on the elimination rate of CFZ but not on the AUC. On the other hand, combined administration of the two HSA NP formulations (CFZ/HSA NP+PTX/HSA NPs) could have resulted in significant influences on both T1/2,initial and AUC0-1h for CFZ compared to those for CFZ/HSA NPs. Fortunately, although administration of CFZ/PTX co-loaded HSA NPs (CFZ/PTX/HSA NPs) led to a lower AUC0-1h than that for CFZ/HSA NPs, a higher AUC0-1h than those for Sb-CFZ and Sb-CFZ+Sb-PTX was observed. This also implies that co-encapsulation of CFZ and PTX in HSA with the simultaneous protection of CFZ and PTX by HSA might minimize drugdrug interactions that existed in the plasma compartment between CFZ and PTX when presented in free forms.

Since a greater difference existed between AUC0-2h and AUC0-24h for all the various formulations of PTX administered as shown in Table 4, pharmacokinetic parameters of T1/2,terminal (h) and AUC0-24h (hg/mL) were selected for comparison. Results in Table 4 indicate that T1/2,terminal (h) and AUC0-24h (hg/mL) for PTX after administration of Sb-PTX were shorter and much higher, respectively, than those for administration of PTX/HSA NPs (7.311.84 vs 12.890.65 min and 6.1712.018 vs 2.5110.497 hg/mL), while they both insignificantly differed from those for administration of Sb-CFZ+Sb-PTX (7.311.84 vs 9.081.92 min and 6.1712.018 vs 7.00402.082 hg/mL). On the other hand, T1/2,terminal (h) and AUC0-24h (hg/mL) for PTX after administration of PTX/HSA NPs greatly differed from those for administration of CFZ/HSA NP+PTX/HSA NPs (12.890.65 vs 13.371.41 min and 2.5110.497 vs 1.9580.824 hg/mL), while they were longer and slightly higher, respectively, than those for administration of CFZ/PTX/HSA NPs (12.890.56 vs 6.540.60 min and 2.5110.497 vs 1.7130.520 hg/mL).

It was reported that the administration of ABI-007 (Nab-paclitaxel or Abraxane) to Sprague-Dawley rats was associated with significantly higher CL and V of PTX compared to Taxol (Sb-PTX) resulting in a shorter T1/2,terminal (h) with a reduction in the AUC0-24h.35 This was attributed to the fact that the initial dilution volume and the central V were higher for PTX formulated as ABI-007 than for PTX formulated as Taxol resulting from Cremophor (as the solubilizing agent used in solvent-based formulations) preventing the distribution of PTX to the circulation and into tissues. What we observed in the comparative pharmacokinetic analysis performed in this study conformed to data in the literature, which showed that T1/2,terminal and AUC0-24h for PTX after administration of Sb-PTX and Sb-CFZ+Sb-PTX were both shorter and much higher, respectively, than those for administration of the albumin-bound counterpart of PTX/HSA NPs and CFZ/HSA NPs+PTX/HSA NPs, while T1/2,terminal and AUC0-24h for PTX after administration of Sb-PTX and PTX/HSA NPs both insignificantly differed from those with combination administration of either solvent-based or albumin-bound counterparts of Sb-CFZ+Sb-PTX and CFZ/HSA NPs+PTX/HSA NPs. Nevertheless, the administration of the co-loaded HSA NP formulation of CFZ/PTX/HSA NPs seemed to result in an even shorter T1/2,terminal (6.540.60 vs 12.890.65, 13.371.41 min) but not increasing AUC0-24h (1.7130.520 vs 2.5110.497, 1.9580.824 hg/mL) for PTX compared to that for administration of PTX/HSA NPs and CFZ/HSA NP+PTX/HSA NPs. The underlying reason for this discrepancy is currently unclear.

The anti-tumor efficacies of drug-loaded HSA NPs were evaluated on MIA Paca-2 cell-xenograft mice. At 14 days after inoculation when tumor volumes had reached 150 mm3, mice were intravenously administered saline, Sb-CFZ, CFZ/HSA NPs, Sb-PTX, PTX/HSA NPs, Sb-CFZ/Sb-PTX (1:2), CFZ/HSA NPs+PTX/HSA NPs (1:2), or CFZ/PTX/HSA NPs. The administration of each formulation was performed on days 0, 1, 7, 8, 14, and 15. Tumor volumes and BWs were assessed three times a week. Tumor growth profiles after administration of the various formulations plotted against time are shown in Figure 5A. TGI (%) compared to the control saline groups was calculated on day 21 after drug administration and on day 46 at termination of the study, and the results are illustrated in Figure 5B. All formulations expressed a greater suppression of tumor growth on both days 21 and 46 than that of saline (2301 mm3). Values of TGI (%) on days 21 and 46 for the Sb-CFZ group showed no improvement compared to the CFZ/HSA NP group, whereas those for the Sb-PTX group showed greater suppression than those for the PTX/HSA NP group on both days 21 and 46. Further, values of TGI (%) on days 21 and 46 for the two combined groups (Sb-CFZ+Sb-PTX and CFZ/HSA NPs+PTX/HSA NPs) all showed increases in TGI (%) compared those of each respective individual group (Sb-CFZ+Sb-PTX vs Sb-CFZ and Sb-PTX and CFZ/HSA NP+PTX/HSA NPs vs CFZ/HSA NPs and PTX/HSA NPs). The results confirmed that a synergic effect on the treatment of MIA PaCa-2 tumors was observed for the combination of CFZ and PTX at a 1:2 ratio regardless of whether Sb-CFZ+Sb-PTZ or CFZ/HSA NP+PTX/HSA NPs were examined. Although only a slight increase in TGI (%) was observed on day 21 but not on day 46 for CFZ/PTX/HSA NPs compared to those for Sb-CFZ+Sb-PTX and CFZ/HSA NPs+PTX/HSA NPs (CFZ/PTX/HSA NPs: 110.20%8.39% Sb-CFZ+Sb-PTX: 89.80%21.19%, CFZ/HSA NP+PTX/HSA NPs: 78.38%16.09%), over 100% of TGI means that tumors had obviously shrunk after treatment with CFZ/PTX/HSA NPs. This seems to indicate that the combination of CFZ and PTX at a 1:2 ratio encapsulated in HSA NPs synergistically improves tumor growth inhibition of MIA PaCa-2 cells.

Figure 5C further shows the weights (g) of tumors excised after tumor-bearing mice were sacrificed on day 46. It clearly shows that, compared to saline (2.330.52 g), there was a significant anti-tumor efficacy with any combined formulations of CFZ and PTX at a 1:2 ratio of Sb-CFZ+Sb-PTX (0.690.12 g), CFZ/HSA NP+PTX/HSA NPs (0.910.33 g), and CFZ/PTX/HSA NPs (0.750.11 g, all p<0.05), but there were no statistically significant differences in tumor weights among the three combined formulations. This further verifies that synergistic improvement in tumor inhibition is achievable with a combination of CFZ and PTX at a 1:2 ratio loaded into solvent-based or HSA NPs. Figure 5D also reveals that the decreases in BWs of mice after administration of various formulations were all smaller than 20% for the 46-day observation period. However, a greater decrease in BW of mice was observed at several time points with the administration of Sb-CFZ+Sb-PTX. This implies that the greater decrease in BW of mice might be attributed to a higher toxicity of solvents used in the solvent-based formulations compared to HSA used in the HSA NP formulations. It could be concluded that the combination therapy of CFZ and PTX at a 1:2 ratio co-loaded in HSA NPs (CFZ/PTX/HSA NP) demonstrated optimal synergistic improvement in the growth inhibition of MIA PaCa-2 cells with less systematic toxicity.

To examine the biodistribution of CFZ and PTX in tumors and major organs, C.B-17 SCID mice bearing MIA CaPa-2 tumors were injected with a single IV dose of various formulations including three solvent-based drugs (Sb-CFZ, Sb-PTX, and Sb-CFZ+Sb-PTX), and four drug-loaded HSA NPs (CFZ/HSA NPs, PTX/HSA NPs, CFZ/HSA NPs+PTX/HSA NPs, and CFZ/PTX/HSA NPs) with respective dosing amounts of CFZ and PTX equivalent to 5 and 10 mg/kg. Tumor tissues and major organs were harvested at 2 or 8 h post-injection, processed to make tissue homogenates, and subsequently analyzed with respect to CFZ and PTX levels by LC-MS/MS, and results are demonstrated in Figure 6. For the biodistribution of CFZ in tumor tissues as shown by Figure 6A (2 h) and 6B (8 h), both Sb-CFZ and CFZ/HSA NP groups presented insignificant difference in CFZ levels, but both displayed significantly higher levels of CFZ than those for the Sb-CFZ+Sb-PTX group at 2 h post-dosing with an accompanying decline in the CFZ level at 8 h post-dosing for those formulations examined. An undetectable CFZ level was seen in tumors for both the CFZ/HSA NP+PTX/HSA NP and CFZ/PTX/HSA NP groups at 2 h post-dosing and for those of the Sb-CFZ+Sb-PTX, CFZ/HSA NP+PTX/HSA NP, and CFZ/PTX/HSA NP groups at 8 h post-dosing. However, the CFZ level in tumors did not seem to be correlated with the tumor growth inhibition rate (TGI %) as revealed by Figure 5B. Similar patterns of CFZ biodistributions in these major organs examined for all formulations as distributed to tumors were observed with predominant distribution to the spleen at 2 h post-dosing with an accompanying decline in the CFZ level at 8 h post-dosing for the formulations examined. Similarly, the least or undetectable CFZ levels were shown in those major organs examined for both the CFZ/HSA NP+PTX/HSA NP and CFZ/PTX/HSA NP groups at 2 and 8 h post-dosing. Since quite lower levels of CFZ (<80 ng/g) were observed in those major organs, systemic toxicity caused by the presence of CFZ might not be highly anticipated.

Figure 6 Tissue distributions of carfilzomib (CFZ) and paclitaxel (PTX) at 2 (A and C) and 8 h (B and D), respectively, after intravenous administration of solvent-based (Sb)-CFZ, Sb-PTX, CFZ/human serum albumin (HSA) nanoparticles (NPs), PTX/HSA NPs, Sb-CFZ+Sb-PTX (1:2), CFZ/HSA NPs+PTX/HSA NPs (1:2), or CFZ/PTX/HSA NPs (equivalent to 5 mg/kg CFZ and 10 mg/kg PTX in each mouse). *p<0.05.

For the biodistribution of PTX in tumors as shown by Figure 6C (2 h) and D (8 h), the Sb-PTX group presented a statistically significantly higher level of PTX than those for the PTX/HSA NP and CFZ/PTX/HSA NP groups at 2 h post-dosing (40.005.91 vs 19.744.51 and 5.722.85 g/g), while those for PTX/HSA NPs and CFZ/HSA NP+PTX/HSA NPs were similar, but both were statistically higher than that for CFZ/PTX/HSA NPs (19.744.51 21.609.54 vs 5.722.85 g/g). However, a slight increase was shown in the PTX level biodistributed in tumors for the Sb-PTX group at 8 h post-dosing compared to that at 2 h post-dosing, whereas those for the remaining formulations at 8 h post-dosing (PTX/HSA NPs, Sb-CFZ+Sb-PTX, CFZ/HSA NPs+PTX/HSA NPs, and CFZ/PTX/HSA NPs) were still statistically significantly lower than that for the Sb-PTX group but showed insignificant differences among them (50.528.60 vs 20.6215.08, 15.066.84, 15.753.88, and 12.983.93 g/g). Similar to CFZ as described above, the PTX level distributed to tumors did not seem to be correlated with the tumor growth inhibition rate (TGI %) as revealed by Figure 5B. Similar patterns of PTX biodistributions in the major organs examined for all formulations as those distributed to tumors were observed with predominant distribution to the liver, spleen, and kidneys at 2 h post-dosing with accompanying significant declines in PTX levels at 8 h post-dosing for those formulations examined. Since quite higher levels of PTX were detected in the liver (134.721.9 g/g at 2 h and 28.9212.62 g/g at 8 h) after administration of Sb-PTX compared to those for the other formulations examined, the higher grade of systemic toxicity caused by administration of Sb-PTX in the presence of this amount of PTX in the liver might be highly expected. It is worth noting that although both 2 and 8 h accumulations of PTX in tumors with the administration of Sb-PTX were 2~3 times more than those for the two combined groups (Sb-CFZ+Sb-PTX and CFZ/HSA NPs+PTX/HSA NPs), TGI (%) values on days 21 and 46 for the two combined groups (Sb-CFZ+Sb-PTX and CFZ/HSA NPs+PTX/HSA NPs) as revealed above all showed increases in TGI (%) compared to that for the Sb-PTX group. This seems to further confirm that the synergistic improvement in tumor growth inhibition is achievable with a combination of CFZ and PTX at a 1:2 ratio loaded into solvent-based or HSA NPs with minimal systemic toxicity.

It was concluded that the effective combination therapy of pancreatic cancer was enabled with treatment of CFZ and PTX co-loaded HSA NPs, which was prepared by a simple one-pot reverse self-assembly method developed in this study. The one-pot reverse self-assembly method was novel and able to optimally prepare HSA NPs loaded with hydrophobic drugs by adjusting the drug/HSA ratio and homogenization process parameters. Without using any hazardous or toxic solvent during preparation of drug-loaded HSA NPs, the one-pot reverse self-assembly method could claim to be environmentally friendly with the ability to co-encapsulate two chemodrugs in HSA NPs with the optimal ratio for synergistic therapy to inhibit tumor growth and minimize systemic toxicity compared to monotherapy. With the related data in this study, it might be able to construct a platform for combination therapy in the future.

This animal experiment was approved by the Institutional Animal Care and Use Committee of Taipei Medical University (Approval No.: LAC-2018-0419) in compliance with the Taiwanese Animal Welfare Act.

This work was supported by the Ministry of Science and Technology, Taiwan, ROC, under grant no. 107-2314-B-038-035-MY3, 108-2314-B-264-001- and 110-2221-E-264-002-.

The authors report no conflicts of interest with respect to this work.

1. American Society of Clinical Oncology. Pancreatic cancer: types of treatment. Internet. Cancer.Net Editorial Board; June 8, 2021. Available from: https://www.cancer.net/cancer-types/pancreatic-cancer/types-treatment. Accessed September 28, 2021.

2. Hidalgo M, Cascinu S, Kleeff J, et al. Addressing the challenges of pancreatic cancer: future directions for improving outcomes. Pancreatology. 2015;15(1):818. doi:10.1016/j.pan.2014.10.001

3. Lei F, Xi X, Batra SK, Bronich TK. Combination therapies and drug delivery platforms in combating pancreatic cancer. J Pharmacol Exp Ther. 2019;370(3):682694. doi:10.1124/jpet.118.255786

4. Wang JP, Wu CY, Yeh YC, et al. Erlotinib is effective in pancreatic cancer with epidermal growth factor receptor mutations: a randomized, open-label, prospective trial. Oncotarget. 2015;6(20):1816218173. doi:10.18632/oncotarget.4216

5. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):16911703. doi:10.1056/NEJMoa1304369

6. Fraunhoffer NA, Abuelafia AM, Bigonnet M, et al. Evidencing a pancreatic Ductal Adenocarcinoma subpopulation sensitive to the proteasome inhibitor Carfilzomib. Clin Cancer Res. 2020;26(20):55065519. doi:10.1158/1078-0432.Ccr-20-1232

7. Kawaguchi K, Igarashi K, Murakami T, et al. MEK inhibitors cobimetinib and trametinib, regressed a gemcitabine-resistant pancreatic-cancer Patient-Derived Orthotopic Xenograft (PDOX). Oncotarget. 2017;8(29):47490. doi:10.18632/oncotarget.17667

8. Roeten MSF, Cloos J, Jansen G. Positioning of proteasome inhibitors in therapy of solid malignancies. Cancer Chemother Pharmacol. 2018;81(2):227243. doi:10.1007/s00280-017-3489-0

9. Naumann K, Schmich K, Jaeger C, Kratz F, Merfort I. Noxa/Mcl-1 balance influences the effect of the proteasome inhibitor MG-132 in combination with anticancer agents in pancreatic cancer cell lines. Anticancer Drugs. 2012;23(6):614626. doi:10.1097/CAD.0b013e3283504e53

10. Papadopoulos KP, Burris HA 3rd, Gordon M, et al. A Phase I/II study of carfilzomib 2-10-min infusion in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2013;72(4):861868. doi:10.1007/s00280-013-2267-x

11. Deshaies RJ. Proteotoxic crisis, the ubiquitin-proteasome system, and cancer therapy. BMC Biol. 2014;12(1):94. doi:10.1186/s12915-014-0094-0

12. Park JE, Chun SE, Reichel D, et al. Polymer micelle formulation for the proteasome inhibitor drug carfilzomib: anticancer efficacy and pharmacokinetic studies in mice. PLoS One. 2017;12(3):e0173247. doi:10.1371/journal.pone.0173247

13. Park JE, Park J, Jun Y, et al. Expanding therapeutic utility of carfilzomib for breast cancer therapy by novel albumin-coated nanocrystal formulation. J Control Release. 2019;302:148159. doi:10.1016/j.jconrel.2019.04.006

14. Peng Q, Zhang S, Yang Q, et al. Preformed albumin Corona, a protective coating for nanoparticles based drug delivery system. Biomaterials. 2013;34(33):85218530. doi:10.1016/j.biomaterials.2013.07.102

15. Sleep D, Cameron J, Evans LR. Albumin as a versatile platform for drug half-life extension. Biochim Biophys Acta. 2013;1830(12):55265534. doi:10.1016/j.bbagen.2013.04.023

16. Park J, Park JE, Hedrick VE, et al. A comparative in vivo study of albumin-coated Paclitaxel Nanocrystals and Abraxane. Small. 2018;14(16):e1703670. doi:10.1002/smll.201703670

17. Stehle G, Sinn H, Wunder A, et al. Plasma protein (albumin) catabolism by the tumor itselfimplications for tumor metabolism and the genesis of cachexia. Crit Rev Oncol Hematol. 1997;26(2):77100. doi:10.1016/s1040-8428(97)00015-2

18. Desai N, Trieu V, Yao Z, et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin Cancer Res. 2006;12(4):13171324. doi:10.1158/1078-0432.Ccr-05-1634

19. Guarneri V, Dieci MV, Conte P. Enhancing intracellular taxane delivery: current role and perspectives of nanoparticle albumin-bound paclitaxel in the treatment of advanced breast cancer. Expert Opin Pharmacother. 2012;13(3):395406. doi:10.1517/14656566.2012.651127

20. Kudarha RR, Sawant KK. Albumin based versatile multifunctional nanocarriers for cancer therapy: fabrication, surface modification, multimodal therapeutics and imaging approaches. Mater Sci Eng C Mater Biol Appl. 2017;81:607626. doi:10.1016/j.msec.2017.08.004

21. Sage H, Johnson C, Bornstein P. Characterization of a novel serum albumin-binding glycoprotein secreted by endothelial cells in culture. J Biol Chem. 1984;259(6):39934007. doi:10.1016/S0021-9258(17)43194-2

22. Hoogenboezem EN, Duvall CL. Harnessing albumin as a carrier for cancer therapies. Adv Drug Deliv Rev. 2018;130:7389. doi:10.1016/j.addr.2018.07.011

23. Han H, Wang J, Chen T, Yin L, Jin Q, Ji J. Enzyme-sensitive gemcitabine conjugated albumin nanoparticles as a versatile theranostic nanoplatform for pancreatic cancer treatment. J Colloid Interface Sci. 2017;507:217224. doi:10.1016/j.jcis.2017.07.047

24. Yu Q, Qiu Y, Li J, et al. Targeting cancer-associated fibroblasts by dual-responsive lipid-albumin nanoparticles to enhance drug perfusion for pancreatic tumor therapy. J Control Release. 2020;321:564575. doi:10.1016/j.jconrel.2020.02.040

25. Yin T, Dong L, Cui B, et al. A toxic organic solvent-free technology for the preparation of PEGylated paclitaxel nanosuspension based on human serum albumin for effective cancer therapy. Int J Nanomedicine. 2015;10:73977412. doi:10.2147/ijn.S92697

26. Yang J, Wang Z, Fang Y, et al. Pharmacokinetics, pharmacodynamics, metabolism, distribution, and excretion of carfilzomib in rats. Drug Metab Dispos. 2011;39(10):18731882. doi:10.1124/dmd.111.039164

27. Chen RF. Removal of fatty acids from serum albumin by charcoal treatment. J Biol Chem. 1967;242(2):173181. doi:10.1016/S0021-9258(19)81445-X

28. Cheng WJ, Chen LC, Ho HO, Lin HL, Sheu MT. Stearyl polyethylenimine complexed with plasmids as the core of human serum albumin nanoparticles noncovalently bound to CRISPR/Cas9 plasmids or siRNA for disrupting or silencing PD-L1 expression for immunotherapy. Int J Nanomedicine. 2018;13:70797094. doi:10.2147/ijn.S181440

29. Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70(2):440446. doi:10.1158/0008-5472.Can-09-1947

30. Chang CE, Hsieh CM, Chen LC, et al. Novel application of pluronic lecithin organogels (PLOs) for local delivery of synergistic combination of docetaxel and cisplatin to improve therapeutic efficacy against ovarian cancer. Drug Deliv. 2018;25(1):632643. doi:10.1080/10717544.2018.1440444

31. Cheng WJ, Lin SY, Chen M, et al. Active tumoral/tumor environmental dual-targeting by non-covalently arming with trispecific antibodies or dual-bispecific antibodies on docetaxel-loaded mPEGylated nanocarriers to enhance chemotherapeutic efficacy and Minimize systemic toxicity. Int J Nanomedicine. 2021;16:40174030. doi:10.2147/ijn.S301237

32. Min JS, Kim J, Kim JH, et al. Quantitative determination of carfilzomib in mouse plasma by liquid chromatography-tandem mass spectrometry and its application to a pharmacokinetic study. J Pharm Biomed Anal. 2017;146:341346. doi:10.1016/j.jpba.2017.08.048

33. Kuo ZK, Lin MW, Lu IH, et al. Antiangiogenic and antihepatocellular carcinoma activities of the Juniperus chinensis extract. BMC Complement Altern Med. 2016;16(1):277. doi:10.1186/s12906-016-1250-6

34. Vlasova I, Saletsky A. Study of the denaturation of human serum albumin by sodium dodecyl sulfate using the intrinsic fluorescence of albumin. J Appl Spectrosc. 2009;76(4):536541. doi:10.1007/s10812-009-9227-6

35. Sparreboom A, Scripture CD, Trieu V, et al. Comparative preclinical and clinical pharmacokinetics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cremophor (Taxol). Clin Cancer Res. 2005;11(11):41364143. doi:10.1158/1078-0432.Ccr-04-2291

36. Ernsting MJ, Murakami M, Undzys E, Aman A, Press B, Li S-D. A docetaxel-carboxymethylcellulose nanoparticle outperforms the approved taxane nanoformulation, Abraxane, in mouse tumor models with significant control of metastases. J Control Release. 2012;162(3):575581. doi:10.1016/j.jconrel.2012.07.043

37. He X, Xiang N, Zhang J, et al. Encapsulation of teniposide into albumin nanoparticles with greatly lowered toxicity and enhanced antitumor activity. Int J Pharm. 2015;487(12):250259. doi:10.1016/j.ijpharm.2015.04.047

38. Kortuem KM, Stewart AK. Carfilzomib. Blood. 2013;121(6):893897. doi:10.1182/blood-2012-10-459883

Continue reading here:
Combination Therapy of Carfilzomib and Paclitaxel for PACs | IJN - Dove Medical Press

3d Cell Culture Market: How Leading Companies Respond to New Market Drivers by Thermo Fisher Scientific, Nanofiber Solutions, Advanced Biomatrix,…

Detailed research added by Adroit Market Research offering a comprehensive analysis of the developments, growth outlook, driving factors, and key players of the 3d Cell Culture market in the latest research report. The research study concisely dissects the 3d Cell Culture and unearths valuable estimations pertaining to the profit projections; market size, sales capacity, and numerous other crucial parameters. Also, the 3d Cell Culture Market report appraises the industry fragments as well as the driving factors impacting the remuneration scale of this industry.

Procure Sample Report + All Related Table and Graphs @https://www.adroitmarketresearch.com/contacts/request-sample/593

For more data or any query mail at[emailprotected]

Competition landscape

-Business Strategies of Leading and prominent market players in 3d Cell Culture.

-Product offering and development analysis.

-Market share & positioning analysis.

-SWOT analysis of the prominent market players.

-Industry-wide business strategies and trends.

Competitive Landscape Thermo Fisher Scientific, Nanofiber Solutions, Advanced Biomatrix, Dickinson and Company

The 3d Cell Culture Market has been segregated into various crucial divisions including applications, types, and regions. Each market segment is intensively studied in the report contemplating its market acceptance, worthiness, demand, and growth prospects. The segmentation analysis will help the client to customize their marketing approach to have a better command of each segment and to identify the most prospective customer base.

Market Analysis by Applications:By Application,(Regenerative medicine,Drug discovery,Stem cell research,Cancer research)

Market Analysis by Type:By End-User, (Academic institutes,Contract research laboratories,Pharmaceutical & biotechnology companies), By Product Type, (Services,Microchips,Bioreactors,Gels,Scaffold-based platforms,Scaffold-free platforms,Solid scaffolds,Nano-porous scaffolds,Micro-porous scaffolds,Macro-porous scaffolds)

1. Which end-user is likely to play a crucial role in the development of the 3d Cell Culture market?

2. Which regional market is expected to dominate the 3d Cell Culture market in 2020?

3. How are consumer trends impacting the operations of market players in the current scenario of the 3d Cell Culture market?

4. Why are market players eyeing opportunities in region 1?

5. What are the growth prospects of the 3d Cell Culture market in region 1 and region 2?

6. What impact does COVID-19 have made on 3d Cell Culture Market Growth & Sizing?

(1) The information presented in the report helps your decision-makers to become prudent and make the best business choices.

(2) The report enables you to see the future of the 3d Cell Culture market and accordingly take decisions that will be in the best interest of your business.

(3) It offers you a forward-looking perspective of the 3d Cell Culture market drivers and how you can secure significant market gains in the near future.

(4) It provides a SWOT analysis of the 3d Cell Culture market along with useful graphics and detailed statistics providing quick information about the markets overall progress throughout the forecast period.

(5) It also assesses the changing competitive dynamics of the 3d Cell Culture market using pin-point evaluation.

6. Impact Analysis of COVID-19 on 3d Cell Culture Market

To conclude, the 3d Cell Culture Market report will provide the clients with a high-yielding market analysis assisting them to understand the market status and come up with new market avenues to capture hold of the market share.

If you have any special requirements, please let us know and we will offer you the report at customized price.

Chapter 1 Market Overview

Chapter 2 Industry Chain

Chapter 3 Environmental Analysis

Chapter 4 Market Segmentation by Type

Chapter 5 Market Segmentation by Application

Chapter 6 Market Segmentation by Region

Chapter 7 Market Competitive

Chapter 8 Major Vendors

Chapter 9 Conclusion

About Us

Contact Us:

Ryan Johnson

Account Manager Global

3131 McKinney Ave Ste 600, Dallas,

TX75204, U.S.A.

Original post:
3d Cell Culture Market: How Leading Companies Respond to New Market Drivers by Thermo Fisher Scientific, Nanofiber Solutions, Advanced Biomatrix,...

Nanomedicine Market to Remain Competitive | Major Giants Continuously Expanding Market The Manomet Current – The Manomet Current

Global Nanomedicine Market is valued at approximately USD 160 billion in 2019 and is anticipated to grow with a healthy growth rate of more than 12.6% over the forecast period 2020-2027. Nanomedicine is one of the most significant applications of nanotechnology used in the treatment, diagnosis, control, and monitoring of biological systems. Nanomedicine utilizes nanoscale manipulation of materials to enhance medicine delivery. Thus, nanomedicine has enabled the treatment alongside various diseases, such as cancer, cardiovascular diseases, and so on. Nanomedicine is the most promising mode of treatment of cancer. This expanding field of medical research can be utilized to discover improved personalized treatment for cancer in the present scenario. With the benefit of the properties of issue at nanoscale, nanomedicine pledges to create innovative drugs with larger efficacy and reduced side-effects than regular therapies. Thus, the surge in prevalence of cancer may act as a major driving factor for the growth of the market all over the world.

According to the National Cancer Institute (NIH), the prevalence of cancer has a major impact on society in the world and across the United States. As of January 2019, around 16.9 million cancer survivors were reported in the United States and is projected to increase to almost 22.2 million by the year 2030. Also, in 2020, an estimated 1,806,590 new cases of cancer were found and will be diagnosed in the United States. Furthermore, the rise in government approvals for the products developed by the manufacturers, along with the increased emergence of newer technologies for drug delivery are the few factors responsible for the high CAGR of the market during the forecast period. For instance, in February 2017, the Celgene International Srl granted approval for its REVLIMID (lenalidomide) from the European Commission, as monotherapy for patients treatment with multiple myeloma. This, in turn, is expected to strengthen the market growth all over the world. However, the high cost of nanomedicine, coupled with strict government norms for product approval are the few major factors inhibiting the market growth over the forecast period of 2020-2027.

Request for a FREE sample of this market research report@ https://www.reportocean.com/industry-verticals/sample-request?report_id=bw2007

The regional analysis of the global Nanomedicine market is considered for the key regions such as Asia Pacific, North America, Europe, Latin America, and the Rest of the World. North America is the leading/significant region across the world in terms of market share owing to the rise in government funding for the nanoscale technology and nanomedicine, and the presence of a significant number of market vendors in the region. Whereas Asia-Pacific is anticipated to exhibit the highest growth rate / CAGR over the forecast period 2020-2027. Factors such as the rise in prevalence of chronic diseases such as cancer, along with the increasing number of venture capital investors in developing countries, such as China and India, would create lucrative growth prospects for the Nanomedicine market across the Asia-Pacific region.

Major market player included in this report are:Abbott LaboratoriesCombiMatrix Corp.General ElectricJohnson & JohnsonMallinckrodt PharmaceuticalsMerck & Co., Inc.Luminex CorporationPfizer Inc.Leadiant Biosciences, Inc.Teva Pharmaceutical Industries Ltd.

The objective of the study is to define market sizes of different segments & countries in recent years and to forecast the values to the coming eight years. The report is designed to incorporate both qualitative and quantitative aspects of the industry within each of the regions and countries involved in the study. Furthermore, the report also caters the detailed information about the crucial aspects such as driving factors & challenges which will define the future growth of the market. Additionally, the report shall also incorporate available opportunities in micro markets for stakeholders to invest along with the detailed analysis of competitive landscape and product offerings of key players. The detailed segments and sub-segment of the market are explained below:

By Modality:DiagnosticsTreatments

By Application:Drug DeliveryVaccinesDiagnostic ImagingRegenerative MedicineImplantsOthers

By Indication:Oncological DiseasesInfectious DiseasesCardiovascular DiseasesOrthopedic DisordersNeurological DiseasesOthers

By Region:North AmericaU.S.CanadaEuropeUKGermanyFranceSpainItalyROE

Asia PacificChinaIndiaJapanAustraliaSouth KoreaRoAPACLatin AmericaBrazilMexicoRest of the World

Furthermore, years considered for the study are as follows:

Historical year 2017, 2018Base year 2019Forecast period 2020 to 2027

Send a request to Report Ocean to understand the structure of the complete report @https://www.reportocean.com/industry-verticals/sample-request?report_id=bw2007

Target Audience of the Global Nanomedicine Market in Market Study:

Key Consulting Companies & AdvisorsLarge, medium-sized, and small enterprisesVenture capitalistsValue-Added Resellers (VARs)Third-party knowledge providersInvestment bankersInvestors

See the original post:
Nanomedicine Market to Remain Competitive | Major Giants Continuously Expanding Market The Manomet Current - The Manomet Current

Medical Physics Market: Growing Incidence of Chronic Diseases in Developing Regions to Drive the Market – BioSpace

Medical Physics Market: Overview

Medical physics has transformed practice of diagnostic and therapeutic medicine, which relates to the use of radiation in medicine. The role of medical physicists in ensuring quality radiation therapy, improving the performance of quality medical imaging is a key factor underpinning the evolution of the medical physics market.

Research in medical physics has focused on assessing the potentially harmful effects of radiation on patients, clinicians, and healthcare staff. Strides that diagnostic and therapeutic medicine has made over the past few years have shaped the growth trajectory of the overall medical physics market. The expanding role of radiology, radiotherapy, and nuclear medicine in diagnostics and therapeutics is a case in point.

Over the years, physicists have been increasingly leaning on discovering processes, procedures, and technologies, that will expand the scope and relevance of healthcare applications. These efforts reinforce the growing outlook of the medical physics market.

Request Brochure of Report - https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=78049

Medical Physics Market: Competitive Analysis and Key Developments

In recent years, medical physicists are being exhorted to improve their contribution to healthcare systems world over. A number of frameworks supporting related strategies is key to offering momentum in this direction. The American Association of Physicists in Medicine (AAPM ) in 2018 devised such a framework Medical Physics 3.0 (MP 3.0) after two years of relentless deliberations to this end. This will help greatly reinvigorate the role of medical physics in patient care in general, expanding the horizon of the market.

The association has urged physicists to securitize their role in medical area, and eventually gain a comprehensive understanding of patient care. Such initiatives are helpful in boosting the prospects of the medical physics market. Experts believe that Medical Physics 3.0 (MP3.0) is likely to set the pace for sustainable excellence in medical physics, my maximizing the contribution of physicists to improvement of human health.

Over the past few years, the medical physics market has been replete with mergers and acquisitions among the healthcare system manufacturers and healthcare providers. This has helped in boosting the adoption of cutting-edge diagnostic imaging in the medical physics market.

Request for Analysis of COVID-19 Impact on Medical Physics Market - https://www.transparencymarketresearch.com/sample/sample.php?flag=covid19&rep_id=78049

Medical Physics Market: Key Trends

Medical physicists have been instrumental in improving the clinical efficacy and safety of wide spectrum of diagnostic and therapeutic modality. These include mammography systems, X-ray systems, computed tomography, magnetic resonance imaging, SPECT, and PET. Key end users include hospitals, academic and research institutes, ambulatory surgery centers, and diagnostic imaging centers.

The need for reducing radiation toxicity in tomotherapy and intensity modulation radiotherapy (IMRT) is boosting the medical physics market. Medical physics is a mix of scientists and healthcare and medical professional. Thus, their role in transforming human and animal health has expanded the vistas in medical physics market.

Request for Custom Research - https://www.transparencymarketresearch.com/sample/sample.php?flag=CR&rep_id=78049

In coming years, future slew of investments in the medical physics market include orthopedics, ophthalmology, medical photonics, surgery, radiogenomics, nano?medicine, dentistry, vascular medicine, and neuro?science.

Medical Physics Market: Regional Analysis

On the regional front, North America and Europe have been vastly attractive medical physics markets. These regional markets have seen the increasing trend of outsourcing of medical physics. In recent years, the role of numerous regional associations, notably in the U.S., in expanding the role of medical physicists in human health has cemented the revenue potential of the global medical physics market. Strides being made by nuclear medicine have spurred revenues in the North America medical physics market.

Pre Book Medical Physics Market Report at https://www.transparencymarketresearch.com/checkout.php?rep_id=78049&ltype=S

The growing incidence of chronic diseases in developing regions such as Asia Pacific and Latin America is opening promising investment scope in these, making them fast emerging markets.

Browse More Trending by Transparency Market Research:

Injectable Benzodiazepine Market: https://www.transparencymarketresearch.com/injectable-benzodiazepine-market.html

Oncology Small Molecule Drugs Market: https://www.transparencymarketresearch.com/oncology-small-molecule-drugs-market.html

Protein A Resin Market: https://www.transparencymarketresearch.com/protein-a-resin-market.html

About Us

Transparency Market Research is a global market intelligence company providing global business information reports and services. Our exclusive blend of quantitative forecasting and trends analysis provides forward-looking insight for several decision makers. Our experienced team of analysts, researchers, and consultants use proprietary data sources and various tools and techniques to gather and analyze information.

Our data repository is continuously updated and revised by a team of research experts so that it always reflects latest trends and information. With a broad research and analysis capability, Transparency Market Research employs rigorous primary and secondary research techniques in developing distinctive data sets and research material for business reports.

ContactTransparency Market Research,90 State Street, Suite 700,Albany, NY 12207Tel: +1-518-618-1030USA - Canada Toll Free: 866-552-3453Website: https://www.transparencymarketresearch.com/

Go here to see the original:
Medical Physics Market: Growing Incidence of Chronic Diseases in Developing Regions to Drive the Market - BioSpace

Nordic Nanovector ASA: Results for the Third Quarter 2020 – BioSpace

OSLO, Norway, Nov. 19, 2020 /PRNewswire/ -- Nordic Nanovector ASA (OSE: NANO) announces its results for the third quarter 2020. A live webcast presentation by Nordic Nanovector's management team will take place today in Oslo at 08.30 CET, see details below. A link to the webcast and the presentation is available from the company's homepage (www.nordicnanovector.com).

Lars Nieba, Interim CEO of Nordic Nanovector, said: "Following the successful interim analysis in August and completion of our private placement in September, we are progressing towards the major value inflection point of three-month top-line data from the PARADIGME clinical study, which is targeted for H2 2021. Generating these data will require us to successfully navigate the latest challenges of increased COVID-19 restrictions. We remain confident in our ability to achieve this goal, aided by the protocol amendments, the possibility to reduce the patient sample, and all the other measures we are actively implementing to drive patient recruitment into PARADIGME."

Q3'2020 Highlights

Events after Q3'2020

Financial Highlights

(Figures in brackets = same period 2019 unless otherwise stated)

Outlook

The company continues to target the readout of three-month top line data from PARADIGME in H2'2021. Approval of protocol amendments is proceeding as planned and completed in the best-recruiting countries, and other initiatives to increase the rate of enrolment are underway. The company also targets the readout of three-month top line data from the second cohort of the Archer-1 trial in H1'2021.

However, the impact of the COVID-19 pandemic on patient recruitment has worsened in light of the emergence of a second wave resulting in severe travel restrictions being implemented in the various countries where we are executing our clinical studies. These restrictions and uncertainty around the duration, severity and geographic scope of the COVID-19 outbreak are projected to slow down the enrolment of patients due to re-prioritisation of hospital activities towards COVID-19 patients and away from clinical studies such as PARADIGME. In addition, travel restrictions could create logistical challenges for the shipment of clinical supplies. Several proactive actions have been taken to minimize the impact of these travel restrictions which could blunt further delays in completing enrolment and delivering preliminary results as targeted.

The company has taken steps to conserve cash and following the recent successful private placement, Nordic Nanovector has a cash runway that extends into Q3'2021.

Despite the challenging times, the many positive actions the company has made in the last nine months have improved the prospects of delivering pivotal results from PARADIGME in H2'2021.

The company continues to believe that, if positive, these trial data could represent a significant value inflection point for the company and its shareholders, confirming Betalutin as a highly promising new targeted therapy that can address the unmet needs of R/R FL patients.

Presentation and live webcast Q3 2020 results

A presentation and live webcast by Nordic Nanovector's management team will take place today at 8:30 am CET.

The webcast can be accessed from http://www.nordicnanovector.com in the section: Investors & Media and a recording will also be available on this page after the event.

The results report and the presentation is available at http://www.nordicnanovector.com in the section: Investors & Media/Reports and Presentation/Interim Reports/2020.

For further information, please contact:

IR enquiries

Malene Brondberg, CFO

Cell: +44 7561 431 762

Email: ir@nordicnanovector.com

Media Enquiries

Mark Swallow/Frazer Hall/David Dible (Citigate Dewe Rogerson)

Tel: +44 203 926 8535

Email: nordicnanovector@citigatedewerogerson.com

About Nordic Nanovector:

Nordic Nanovector is committed to develop and deliver innovative therapies to patients to address major unmet medical needs and advance cancer care. The Company aspires to become a leader in the development of targeted therapies for haematological cancers. Nordic Nanovector's lead clinical-stage candidate is Betalutin, a novel CD37-targeting antibody-radionuclide-conjugate designed to advance the treatment of non-Hodgkin's lymphoma (NHL). NHL is an indication with substantial unmet medical need, representing a growing market forecast to be worth nearly USD 29 billion by 2026. Nordic Nanovector retains global marketing rights to Betalutin and intends to actively participate in the commercialisation of Betalutin in the US and other major markets.

Further information can be found at http://www.nordicnanovector.com .

Forward-looking statements

This press release contains certain forward-looking statements. These statements are based on management's current expectations and are subject to uncertainty and changes in circumstances, since they relate to events and depend on circumstances that will occur in the future and which, by their nature, will have an impact on Nordic Nanovector's business, financial condition and results of operations. The terms "anticipates", "assumes", "believes", "can", "could", "estimates", "expects", "forecasts", "intends", "may", "might", "plans", "should", "projects", "targets", "will", "would" or, in each case, their negative, or other variations or comparable terminology are used to identify forward-looking statements. These forward-looking statements are not historic facts. There are a number of factors that could cause actual results and developments to differ materially from those expressed or implied in the forward-looking statements. Factors that could cause these differences include, but are not limited to, risks associated with implementation of Nordic Nanovector's strategy, risks and uncertainties associated with the development and/or approval of Nordic Nanovector's product candidates, ongoing and future clinical trials and expected trial results, the ability to commercialise Betalutin, technology changes and new products in Nordic Nanovector's potential market and industry, Nordic Nanovector's freedom to operate (competitors patents) in respect of the products it develops, the ability to develop new products and enhance existing products, the impact of competition, changes in general economy and industry conditions, and legislative, regulatory and political factors. No assurance can be given that such expectations will prove to have been correct. Nordic Nanovector disclaims any obligation to update or revise any forward-looking statements, whether as a result of new information, future events or otherwise.

This information is subject to a duty of disclosure pursuant to Sections 4-2 and 5-12 of the Securities Trading Act.

This information was brought to you by Cision http://news.cision.com

https://news.cision.com/nordic-nanovector/r/nordic-nanovector-asa--results-for-the-third-quarter-2020,c3239830

The following files are available for download:

View original content:http://www.prnewswire.com/news-releases/nordic-nanovector-asa-results-for-the-third-quarter-2020-301176750.html

SOURCE Nordic Nanovector

Company Codes: Bloomberg:NANO@NO, ISIN:NO0010597883, Oslo:NANO, RICS:NANO.OL

Visit link:
Nordic Nanovector ASA: Results for the Third Quarter 2020 - BioSpace

Healthcare Nanotechnology (Nanomedicine) Market is Slated To Grow Rapidly In The Coming Years (2020 2028) – TechnoWeekly

The global Healthcare Nanotechnology (Nanomedicine) Market study offers a compilation of the current, historical, and future outlook of the industry as well as the factors responsible for market growth. With a SWOT analysis, the business study highlights the weaknesses, strengths, opportunities, and threats of each Healthcare Nanotechnology (Nanomedicine) Market player in a comprehensive way.

Sample Copy of This Report: https://www.quincemarketinsights.com/request-sample-64780?utm_source=SA/technoweekly

Under the COVID-19 outbreak analysis, this report provides analysis of import, supply chain, and export to future influence on the industry and regional government policies. Enterprise competition pattern, detailed analysis about market status, advantages and disadvantages of enterprise products, macroeconomic policies and regional industrial layout characteristics, industry development trends have also been included. The trends of product sales channel will be offered as well.

Considering COVID-19, this report offers a complete and exhaustive analysis on how the epidemic has pushed transformation and reform in the industry. The market study can help understand the market expansion and strategies for business accordingly. In the strategy analysis, it gives insights from market positioning marketing channel to potential growth strategies, thereby providing an in-depth analysis for new entrants or existing competitors in the Healthcare Nanotechnology (Nanomedicine) industry.

Key Players: Sanofi SA, Pfizer Inc., Celgene Corporation, Luminex Corporation, and Taiwan Liposome Company Ltd..

The market analysis on the Healthcare Nanotechnology (Nanomedicine) offers a birds eye view of the current proceeding within the Healthcare Nanotechnology (Nanomedicine) Market. The market analysis report has incorporated an analysis of various factors that augment the markets growth. It constitutes restraints, trends, and drivers that transform the market in either a negative orpositive manner.

Healthcare Nanotechnology (Nanomedicine) Market

Get ToC for the overview of the premium report @ https://www.quincemarketinsights.com/request-toc-64780?utm_source=SA/technoweekly

This market analysis also offers the scope of different segments and applications that can actually impact the market in the future. The thorough information is based on present trends and historic milestones. The market analysis mentions the volume of sales by region from 2015 to 2026. A thorough evaluation of the limitations included in the market analysis portrays the drivers and gives room for strategic planning.

Global Healthcare Nanotechnology (Nanomedicine) Market: Segment Analysis

Each type offers information about return sover the forecast period of 2018 to 2028. The sales method segment also offers revenue by volume and sales over the forecast period of 2018 to 2028. Understanding the segments helps in classifying the importance of various factors that support market growth.

Following are the segmentation covered by the market study: By Application (Drug Delivery, Biomaterials, Active Implants, Diagnostic Imaging, Tissue Regeneration), By Disease (Cardiovascular Diseases, Oncological Diseases, Neurological Diseases, Orthopedic Diseases, Infectious Diseases and Other Diseases)

Global Healthcare Nanotechnology (Nanomedicine) Market: Regional Analysis

The market analysis report includes a thorough study of various factors that determine regional growth such as environmental, economic, social political status, technology, and region. Market analysis has studied the data of sales, revenue, and manufacturers of each region. The market analysis provides region-wise volume and revenue for the forecast period of 2016 to 2028. This market analysis will support the market participants to understand the potential worth of investment in a particular region.

Region- wise Analysis of the Global Healthcare Nanotechnology (Nanomedicine) Market covers: North America Europe Asia Pacific Middle East and Africa South America

Make an Enquiry for purchasing this Report @ https://www.quincemarketinsights.com/enquiry-before-buying-64780?utm_source=SA/AF

Global Healthcare Nanotechnology (Nanomedicine) Market: Competitive Landscape

This market analysis report classifies numerous key manufacturers of the industry. It supports the reader in understanding the policies and collaborations that the industry participants are focusing on in order to combat competition in the industry. The comprehensive market analysis offers a note worthy microscopic look at the industry. The market analysis can classify the footprints of the manufacturers by giving the global revenue of manufacturers and sales by manufacturers, and the global price of manufacturers over the forecast period of 2018 to 2028.

Major Aspects covered in the Report are

Further, the report provides niche insights for a decision about every possible segment, helping in the strategic decision-making process and market size estimation of the Healthcare Nanotechnology (Nanomedicine) Market on a regional and global basis. Unique research designed for market size estimation and forecast is used for the identification of major companies operating in the market with related developments. The report has an exhaustive scope to cover all the possible segments, helping every stakeholder in the Healthcare Nanotechnology (Nanomedicine) Market.

Reasons for buying this report:

Why Do Companies Trust QMI?

ABOUT US:

QMI has the most comprehensive collection of market research products and services available on the web. We deliver reports from virtually all major publications and refresh our list regularly to provide you with immediate online access to the worlds most extensive and up-to-date archive of professional insights into global markets, companies, goods, and patterns.

Contact:

Quince Market Insights

Office No- A109

Pune, Maharashtra 411028

Phone: APAC +91 706 672 4848 / US +1 208 405 2835 / UK +44 1444 39 0986

Email: [emailprotected]

Web: https://www.quincemarketinsights.com

Excerpt from:
Healthcare Nanotechnology (Nanomedicine) Market is Slated To Grow Rapidly In The Coming Years (2020 2028) - TechnoWeekly

Kanazawa University research: The Nano Life Science Institute (WPI-NanoLSI) at Kanazawa University is organizing the 4th NanoLSI Symposium on 26 and…

KANAZAWA, Japan, Nov. 17, 2020 /PRNewswire/ --The 4th NanoLSI Symposium will be a virtual meeting covering bio-imaging, sensing, and manipulation for medical science. The Nano Life Science Institute (WPI-NanoLSI) was established in 2017 as part of the World Premier International Research Center Initiative of the Ministry of Education, Culture, Sports, Science and Technology.

Symposium website

https://nanolsi.kanazawa-u.ac.jp/en/4th-sympo/

Flyer

https://nanolsi.kanazawa-u.ac.jp/wp-content/uploads/2020/11/NanoLSI_symposium_2020.jpg

Program details

https://nanolsi.kanazawa-u.ac.jp/wp-content/uploads/2020/11/program.pdf

Nano Life Science Institute (WPI-NanoLSI) aims to acquire a fundamental understanding of various life phenomena, such as development, disease, and aging at the nano-level through transdisciplinary research of Nanometrology, Life Science, Supramolecular Chemistry, and Computational Science.

To further develop interdisciplinary research, the WPI-NanoLSI has organized annual international symposia to build new collaborative relationships with researchers across the world.

This year's fourth symposium will be held online because of restrictions to travel due to the COVID-19 pandemic. To facilitate research exchange that is only possible through online, the WPI-NanoLSI has invited prominent researchers from Europe, North America, and Asia to give talks during the symposium, where each session is scheduled according to the local times of the speakers.

This is an excellent opportunity to gain valuable insights into the latest research findings in bio-imaging, sensing, and manipulation for medical science.

Details

Date: 26 -27 November 2020

Format: Virtual conference (via Zoom) that is free of charge

Timetable

Thursday 26 November

07:40 07:50 (JST) Opening remarks

08:00 10:00 (JST) 1st session

Molecular and cellular dynamics in biological regulation and regenerative medicine

Chair: Prof. Kunio MATSUMOTO/ NanoLSI/CRI

Speakers: Prof. Simon SCHEURING/ Weill Cornell Medicine

Assoc. Prof. Yusuke MIYANARI/ NanoLSI/CRI

Prof. Kenneth S. Zaret/ University of Pennsylvania

Prof. Takanori TAKEBE/ Tokyo Medical and Dental University

10:00 11:00 (JST). Virtual poster session

*Need Separate registration. Please see website

13:30 15:30 (JST) 2nd session

Chemistry-Driven Challenges: from Molecule to Nano/Microscale

Chair: Assoc.Prof. Satoshi ARAI/ NanoLSI

Speakers: Prof. Tomoki OGOSHI/ Kyoto University

Assoc. Prof. Toshinori FUJIE/ Tokyo Institute of Technology

Assoc. Prof. Etsuo SUSAKI/ The University of Tokyo

Prof. Susumu KITAGAWA/ Kyoto University

15:30 16:00 (JST) Video session #1

17:00 19:00 (JST) 3rd session

Nano-scale approaches to physiological and pathological phenomena

Chair: Prof. Toshio ANDO/ NanoLSI

Speakers: Dr. Nobuo NODA/ Institute of Microbial Chemistry

Prof. Noriyuki KODERA/ NanoLSI

Assoc. Prof. Shinji WATANABE/ NanoLSI

Prof. Victor SHAHIN/ University of Mnster

19:00 19:30 (JST) Video session #2

Friday 27 November

10:00 12:00 (JST) 4th session

Imaging approaches to explore cancer biology

Chair: Prof. Masanobu OSHIMA/ NanoLSI/CRI

Speakers: Asst. Prof. Keehoon JUNG/Seoul National University College of Medicine

Prof. Takeshi IMAMURA/ Ehime University

Asst. Prof. Ann-Marie CHACKO/ Duke-NUS Medical School

Prof. Masanobu OSHIMA/ NanoLSI/CRI

12:00 12:05 (JST) Closing remarks

Further information

About WPI nanoLSI Kanazawa University Hiroe YonedaVice Director of Public AffairsWPI Nano Life Science Institute (WPI-NanoLSI)Kanazawa UniversityKakuma-machi, Kanazawa 920-1192, JapanEmail: nanolsi-office@adm.kanazawa-u.ac.jpTel: +81 (76) 234-4550

About Nano Life Science Institute (WPI-NanoLSI)

https://nanolsi.kanazawa-u.ac.jp/en/

Nano Life Science Institute (NanoLSI), Kanazawa University is a research center established in 2017 as part of the World Premier International Research Center Initiative of the Ministry of Education, Culture, Sports, Science and Technology. The objective of this initiative is to form world-tier research centers. NanoLSI combines the foremost knowledge of bio-scanning probe microscopy to establish 'nano-endoscopic techniques' to directly image, analyze, and manipulate biomolecules for insights into mechanisms governing life phenomena such as diseases.

About Kanazawa University

http://www.kanazawa-u.ac.jp/e/

As the leading comprehensive university on the Sea of Japan coast, Kanazawa University has contributed greatly to higher education and academic research in Japan since it was founded in 1949. The University has three colleges and 17 schools offering courses in subjects that include medicine, computer engineering, and humanities.

The University is located on the coast of the Sea of Japan in Kanazawa a city rich in history and culture. The city of Kanazawa has a highly respected intellectual profile since the time of the fiefdom (1598-1867). Kanazawa University is divided into two main campuses: Kakuma and Takaramachi for its approximately 10,200 students including 600 from overseas.

SOURCE Kanazawa University

See original here:
Kanazawa University research: The Nano Life Science Institute (WPI-NanoLSI) at Kanazawa University is organizing the 4th NanoLSI Symposium on 26 and...

Antibacterial Properties of OsteoFab PEKK Implants Defined in New Research, Outperforming PEEK and Titanium – PR Web

We have been continually impressed with the strong bacteria resistance and pro-bone cell responses of OPMs PEKK samples, said Dr. Thomas Webster of Northeastern University. All resulting from optimal surface properties without resorting to the use of antibiotics or the release of growth factors

SOUTH WINDSOR, Conn. (PRWEB) November 18, 2020

Oxford Performance Materials, Inc. (OPM), an industry leader in advanced materials science and high performance additive manufacturing (HPAM), today announced additional results of an ongoing scientific study analyzing the antibacterial properties of 3D printed PEKK (poly-ether-ketone-ketone). These new results definitively establish a mechanism of action for how 3D printed PEKK exhibits inherent antibacterial characteristics, further confirming PEKKs role in combatting implant infections. In a continued partnership with Dr. Thomas Webster of Northeastern University, this research builds upon a September 2017 study(1) where samples produced by OPMs proprietary OsteoFab process demonstrated for the first time the promise that nanostructured PEKK has for numerous anti-infection orthopedic implant applications.

In a second set of experiments conducted by Dr. Webster in December 2019, it was determined that 3D printed PEKK demonstrated significant reductions for all bacteria colonization(2) when measured via colony forming units, crystal violet staining, and live/dead assays. This was true for all tested bacteria strains, which included S. epidermidis, P. aeruginosa, and MRSA, and all tested materials, as 3D printed PEKK significantly outperformed commercially available titanium and PEEK controls. This study was important for two primary reasons: it confirmed the results seen in the 2017 study(1) in the presence of titanium and PEEK controls and it raised the question of why PEKK is superior in this aspect, hinting that 3D printed PEKK may have a biologically optimal surface energy in the context of bacterial aversion.

In April of 2020, a follow-up study was conducted to determine the mechanisms of the antibacterial properties of 3D printed PEKK after 24 hours of culture, specifically examining protein adsorption and correlating that adsorption to bacteria response. Based on the test results(2), 3D printed PEKK exhibited a surface energy (35.7 mN/m) that was much closer to proteins lubricin (40 mN/m), mucin (42-46 mN/m), and casein (48 mN/m), which are all proteins known to reduce bacteria attachment and colonization naturally in vivo. When compared to the surface energies of PEEK (16.3 mN/m) and titanium (62.5 mN/m), it was evident 3D printed PEKK is optimal in terms of similarity to these naturally occurring proteins. As expected, the experiment showed greater protein adsorption of lubricin, mucin, and casein on PEKK in comparison to PEEK and titanium controls.

This greater protein adsorption was then correlated to greater bacteria inhibition of 3D printed PEKK compared to commercially available PEEK and titanium. For this portion of the study, samples were coated with lubricin, mucin, and casein, and bacteria attachment was characterized using colony forming unit counts, crystal violet staining, and live/dead assays. All results correlated the increased protein adsorption to decreased bacteria colonization for S. epidermidis, P. aeruginosa, and MRSA, again compared to PEEK and titanium controls.

This finding is extremely important as it provides objective evidence explaining why OPMs 3D printed PEKK exhibits inherent antibacterial effects. We have been continually impressed with the strong bacteria resistance and pro-bone cell responses of OPMs PEKK samples, said Dr. Webster. All resulting from optimal surface properties without resorting to the use of antibiotics or the release of growth factors, as our studies have shown. Equally significant are the implications of lubricin adsorption onto 3D printed PEKK, in terms of future joint and cartilage applications of the OsteoFab technology platform. Lubricin is a naturally occurring protein that is secreted in synovial joints; it coats surrounding cartilage and contributes to the overall integrity of the joint. With demonstrated adhesion to 3D printed PEKK, new devices could provide even more benefits in an increasing number of orthopedic applications.

The antibacterial attributes of 3D printed PEKK are significant since they address a key area of growing concern in medicine orthopedic implant infections. 3D printed PEKKs antibacterial properties, as detailed in this new study, will provide another important layer of differentiation for the performance of OsteoFab medical devices in the marketplace. Results from these experiments will be available in a forthcoming publication.

About Oxford Performance Materials, Inc.

Oxford Performance Materials, Inc. was founded in 2000 to exploit and commercialize the worlds highest performing thermoplastic, PEKK (poly-ether-ketone-ketone). OPMs Materials business has developed a range of proprietary, patented technologies for the synthesis and modification of a range of PAEK polymers that are sold under its OXPEKK brand for biomedical and industrial applications. The Company is a pioneer in 3D printing. OPM Biomedicals OsteoFab technology is in commercial production in numerous orthopedic implant applications, including cranial, facial, spinal, and sports medicine devices. OPM is the first and only company to receive FDA 510(k) clearance to manufacture 3D printed, patient-specific polymeric implants, and the company has six 510(k) clearances in its portfolio. OPM Industrial produces 3D printed OXFAB production parts for highly demanding applications in the energy, transportation, and semiconductor markets. OXFAB structures offer significant weight, cost, and time-to-market reductions that are defined in a set of specified performance attributes in the exhaustive OPM B-Basis database, developed in conjunction with NASA. For more information, please visit: http://www.oxfordpm.com

References

Antibacterial Properties of PEKK for Orthopedic Applications, International Journal of Nanomedicine, Dovepress 15-Sep-2017, Mian Wang, Garima Bhardwaj (Department of Chemical Engineering, Northeastern University, Boston MA) and Thomas J. Webster (Department of Chemical Engineering, Northeastern University, Boston MA and Wenzhou Institute of Biomaterials and Engineering, Wenzhou Medical University, Wenzhou, China). The full study by Mian Wang, et al. may be viewed at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5592909/

2. https://oxfordpm.com/uploads/files/OPM-T._Webster_AM_Med_Summit_19Oct2020_website.pdf?v=1605303394194

Company Contact: Bernie Plishtin Oxford Performance Materials 860-656-9446 bplishtin@oxfordpm.com

Share article on social media or email:

Originally posted here:
Antibacterial Properties of OsteoFab PEKK Implants Defined in New Research, Outperforming PEEK and Titanium - PR Web

Messenger RNA the Therapy of the Future – Newswise

With two companies leading the way in development of an mRNA (messenger RNA) vaccine to ward off COVID-19, one Houston Methodist physician researcher sees it as the first step for what has the potential to become the therapy of the future for all types of diseases. John Cooke, MD, PhD, director of the Research Institutes RNA therapeutics program, says mRNA is basically writing biological code to tell our bodies what proteins to make. Scientists need only the genomic sequence of the virus to come up with strategies to intervene.

Most vaccines today are still viral vaccines they are inactivated virus, so its potentially infectious and you have to have virus on hand, Cooke said. With mRNA, youre just writing code which is going to tell the cell to make a viral protein one part of a viral protein to stimulate an immune response. And, heres the wonderful thing, you dont even need the virusin hand, just its DNA code. His team is working with Houston Methodists nanomedicine team to develop better delivery options for mRNA, and he sees its potential in many fields: cancer, cardiovascular disease, liver disease, other infectious diseases and even rejuvenation.

Dr. John Cooke is available for interviews to discuss the process and promise of mRNA technology.

More:
Messenger RNA the Therapy of the Future - Newswise

SHAREHOLDER ALERT: WeissLaw LLP Reminds WTRE, TOTA, HCAC, and OAC Shareholders About Its Ongoing Investigations – PRNewswire

NEW YORK, Nov. 16, 2020 /PRNewswire/ --

If you own shares in any of the companies listed above and would like to discuss our investigations or have any questions concerning this notice or your rights or interests, please contact:

Joshua Rubin, Esq.WeissLaw LLP1500 Broadway, 16th FloorNew York, NY 10036(212)682-3025(888) 593-4771[emailprotected]

Watford Holdings Ltd. (NASDAQ: WTRE)

WeissLaw LLPis investigating possible breaches of fiduciary duty and other violations of law by the board of directors of Watford Holdings Ltd. (NASDAQ: WTRE) in connection withthe proposed acquisition of the company by Arch Capital Group Ltd. ("ACGL"). Under the terms of the acquisition agreement, WTRE shareholders will be entitled to receive $31.10 in cash for each share of WTRE common stock that they own. If you own WTRE shares and wish to discuss this investigation or your rights, please call us at one of the numbers listed above or visit our website: https://weisslawllp.com/wtre/

Tottenham Acquisition I Limited (NASDAQ: TOTA)

WeissLaw LLP is investigating possible breaches of fiduciary duty and other violations of law by the board of directors ofTottenham Acquisition I Limited (NASDAQ: TOTA) in connection with the company's proposed merger with privately-held clinical-stage biopharmaceutical company, Clene Nanomedicine, Inc. ("Clene"). Under the terms of the merger agreement, TOTA will acquire Clene through a reverse merger that will result in Clene becoming a public company listed on the Nasdaq Capital Market. The proposed transaction values Clene at $542.5 million. If you own TOTA shares and wish to discuss this investigation or your rights, please call us at one of the numbers listed above or visit our website: http://www.weisslawllp.com/tottenham-acquisition-i-limited/

Hennessy Capital Acquisition Corp. IV(NASDAQ: HCAC)

WeissLaw LLP is investigating possible breaches of fiduciary duty and other violations of law by the board of directors of Hennessy Capital Acquisition Corp. IV(NASDAQ: HCAC)in connection with HCAC's merger with privately-held Canoo Holdings Ltd. ("Canoo"). Under the terms of the proposed transaction, HCAC will acquire Canoo through a reverse merger that will result in Canoo becoming a publicly-listed company. If you own HCAC shares and wish to discuss this investigation or your rights, please call us at one of the numbers listed above or visit our website: http://www.weisslawllp.com/hennessy-capital-acquisition-corp-iv/

Oaktree Acquisition Corp. (NYSE: OAC)

WeissLaw LLP is investigating possible breaches of fiduciary duty and other violations of law by the board of directors of Oaktree Acquisition Corp. (NYSE: OAC) in connection with the company's proposed merger with privately-held telehealth company Hims, Inc. ("Hims"). Under the terms of the merger agreement, OAC will acquire Hims through a reverse merger that will result in Hims becoming a publicly-listed company on the New York Stock Exchange under the new ticker symbol "HIMS." If you own OAC shares and wish to discuss this investigation or your rights, please call us at one of the numbers listed above or visit our website: https://www.weisslawllp.com/oac/

SOURCE WeissLaw LLP

http://weisslawllp.com

Originally posted here:
SHAREHOLDER ALERT: WeissLaw LLP Reminds WTRE, TOTA, HCAC, and OAC Shareholders About Its Ongoing Investigations - PRNewswire

Bio-convergence may be next big thing from Israel – Cleveland Jewish News

Testing new cancer drugs on 3D-printed tumor cells. Analyzing the gut microbiome to determine the best medicine for Crohns disease. Identifying genetic pairs so that a drug can turn off a cancer cell.

These are just a few of the overlaps of biology, physics, computer science, mathematics, engineering, materials science and nanotechnology that make up the new field of bio-convergence.

Testing new cancer drugs on 3D-printed tumor cells. Analyzing the gut microbiome to determine the best medicine for Crohns disease. Identifying genetic pairs so that a drug can turn off a cancer cell.

These are just a few of the overlaps of biology, physics, computer science, mathematics, engineering, materials science and nanotechnology that make up the new field of bio-convergence.

The Israel Innovation Authority is betting on bio-convergence as the next big thing from the startup nation.

The IIA announced in January that it would invest up to about $30 million in bio-convergence technologies and companies. In July, the governmental agency added a call for R&D proposals from academia and industry in the field of bio-convergence with a budget of $4 million.

In August, a third call for bio-convergence proposals this time for collaboration between Israeli and South Korean companies came with a $4.16 million budget and a plan to provide grants up to 50 percent of R&D expenses.

The bio-convergence push is a bit of a departure for the IIA.

We usually work in a bottom up approach and very seldom intervene and select a specific technological field for strategic investments, Anya Eldan, vice president of the IIAs Startup Division, said. In the past it has been cyber and fintech. Now weve identified bio-convergence as the next growth engine for the Israeli economy.

Developing disease-fighting drugs the old-fashioned way takes decades and billions of dollars. The new era of pharma will see researchers from multiple disciplines working together.

When we started to look for the next growth engine, we realized that none of the major pharmaceutical companies are invested in bio-convergence, Eldan explained. Theyre looking at biology but theyre not sure how to approach it.

Israel, however, is a small country where everyone knows one another, making it relatively easy to put together multidisciplinary teams.

On a folksier level, Eldan said Israelis large Friday night Shabbat dinners naturally foster collaboration.

People have to find things to talk about. So, a doctor mentions at the dinner table he has a patient without a solution and the engineer in the family says, That shouldnt be, and from there a startup is born.

This trend could have a major impact on the global health care system crisis. Health expenditures worldwide were expected to reach $10 trillion by 2022 even before the advent of COVID-19. Half the U.S. population is considered chronically ill, accounting for some 85% of the total expenditure on healthcare services.

The IIA has identified approximately 80 companies working in the field of bio-convergence. Much of the knowhow is tucked away in Israels academic centers.

Ronit Satchi Fainaro is a prime example.

Named Woman of the Year by the Israeli business publication Globes in 2019, Satchi Fainaro heads the 30-person cancer research and nano-medicine laboratory in the department of physiology and pharmacology at Tel Aviv University.

Satchi Fainaros lab includes research associates in biology, chemistry, medical engineering, bioinformatics and even an architecture student, she said. We are living in the post-genomic era, so theres a lot of room for computer science.

Satchi Fainaros lab developed a method for 3D printing cancerous brain tumors using MRI on images.

We do the image analysis and convert it to a file that a 3D printer can read. Then we create a 3D-printed tumor its like a Mini-Me of that tumor, she said, referring to the Mike Meyers character from the Austin Powers films.

Satchi Fainaros lab creates not one but up to 20 mini-tumors, which are then connected to a set of tubes and pumps that deliver simulated blood and most importantly chemotherapy or other cancer treatments. That allows physicians to test drugs on a perfect copy of the actual tumor.

Satchi Fainaro is also working on an immunotherapy nano-vaccine for melanoma and an immune-modulated response against Covid-19.

Jonathan Solomon and Assaf Oron are the CEO and chief business development officer, respectively, of BiomX, another Israeli bio-convergence pioneer.

BiomX is working on a treatment for Crohns and other inflammatory bowel diseases by sequencing the bacteria in a patients microbiome in order to deliver a gut-punch to the exact bacteria causing symptoms.

BiomXs technology is based on the hypothesis that these diseases are driven by the microbiome and that specific bacteria seem to be pro-inflammatory and antibiotic resistant, Solomon tells ISRAEL21.

Bacteriophage image courtesy of BiomX

Bio-convergence techniques assure that a patient receives the right phages a type of virus that infects bacteria to kill only the offending microbes.

Antibiotics are very indiscriminate killers, Solomon said, which leads to the killing of both harmful and beneficial bacteria and, in many cases, the development of antibiotic-resistant bacteria. Phages are very precise. There are no side effects.

Beyond IBD, acne and liver disease are next on BiomXs list. Even cancer is a possibility.

BiomXs technology was initially developed and licensed from the Weizmann Institute in Rehovot. The company has 100 employees and is publicly traded in the United States.

Then theres Pangea, whose bio-convergence niche is synthetic lethality.

Its a very simple notion with a confusing name, chief technology officer Tuvik Beker said.

The basic idea is that genes often act in pairs. If a gene is acting unnaturally in a tumor (for example, it may be shut down due to mutation or abnormal expression), identifying and shutting down the genes partner can selectively kill the malignant cells.

Pangea brings the computational element.

If there are 20,000 genes in the human genome, simple math means there are about 400 million gene pairs, Beker explained. To test all these pairs for synthetic lethality is very challenging.

As a result, most companies restrict themselves to a couple of hundred genes that they know are important in cancer. Pangea, by contrast, looks for paired genes that are vulnerable to collateral damage, not just the ones driving the tumor, Beker said.

The Tel Aviv-based company hopes to launch its service commercially as early as 2021 with a focus on providing personalized treatment recommendations.

See the original post here:
Bio-convergence may be next big thing from Israel - Cleveland Jewish News

Nanomedicine Market: Clear Understanding of The Competitive Landscape and Key Product Segments – PRnews Leader

Overview:

Nanomedicineis an offshoot of nanotechnology, and refers to highly-specific medical intervention at the molecular scale for curing diseases or repairing damaged tissues. Nanomedicine uses nano-sized tools for the diagnosis, prevention and treatment of disease, and to gain increased understanding of the complex underlying pathophysiology of the disease. It involves three nanotechnology areas of diagnosis, imaging agents, and drug delivery with nanoparticles in the 11,000 nm range, biochips, and polymer therapeutics.

Click Here to Get Sample Premium Report @https://www.trendsmarketresearch.com/report/sample/9807

Majority of nanomedicines prescribedcurrently, allow oral drug delivery and its demand is increasing significantly. Although these nanovectors are designed to translocate across the gastrointestinal tract, lung, and bloodbrain barrier, the amount of drug transferred to the organ is lower than 1%; therefore improvements are challenging. Nanomedicines are designed to maximize the benefit/risk ratio, and their toxicity must be evaluated not only by sufficiently long term in vitro and in vivo studies, but also pass multiple clinical studies.

Market Analysis:

The Global Nanomedicine Market is estimated to witness a CAGR of 17.1% during the forecast period 20172023. The nanomedicine market is analyzed based on two segments therapeutic applications and regions.

The major drivers of the nanomedicine market include its application in various therapeutic areas, increasing R&D studies about nanorobots in this segment, and significant investments in clinical trials by the government as well as private sector. The Oncology segment is the major therapeutic area for nanomedicine application, which comprised more than 35% of the total market share in 2016. A major focus in this segment is expected to drive the growth of the nanomedicine market in the future.

Regional Analysis:

The regions covered in the report are the Americas, Europe, Asia Pacific, and Rest of the World (ROW). The Americas is set to be the leading region for the nanomedicine market growth followed by Europe. The Asia Pacific and ROW are set to be the emerging regions. Japan is set to be the most attractive destination and in Africa, the popularity and the usage of various nano-drugs are expected to increase in the coming years. The major countries covered in this report are the US, Germany, Japan, and Others.

Therapeutic Application Analysis:

Nanomedicines are used as fluorescent markers for diagnostic and screening purposes. Moreover, nanomedicines are introducing new therapeutic opportunities for a large number of agents that cannot be used effectively as conventional oral formulations due to poor bioavailability. The therapeutic areas for nanomedicine application are Oncology, Cardiovascular, Neurology, Anti-inflammatory, Anti-infectives, and various other areas. Globally, the industry players are focusing significantly on R&D to gain approval for various clinical trials for future nano-drugs to be commercially available in the market. The FDA should be relatively prepared for some of the earliest and most basic applications of nanomedicine in areas such as gene therapy and tissue engineering. The more advanced applications of nanomedicine will pose unique challenges in terms of classification and maintenance of scientific expertise.

Key Players:

Merck & Co. Inc., Hoffmann-La Roche Ltd., Gilead Sciences Inc., Novartis AG, Amgen Inc., Pfizer Inc., Eli Lilly and Company, Sanofi, Nanobiotix SA, UCB SA and other predominate & niche players.

Competitive Analysis:

At present, the nanomedicine market is at a nascent stage but, a lot of new players are entering the market as it holds huge business opportunities. Especially, big players along with the collaboration with other SMBs for clinical trials of nanoparticles and compounds are coming with new commercial targeted drugs in the market and they are expecting a double-digit growth in the upcoming years. Significant investments in R&D in this market are expected to increase and collaborations, merger & acquisition activities are expected to continue.

Benefits:

The report provides complete details about the usage and adoption rate of nanomedicines in various therapeutic verticals and regions. With that, key stakeholders can know about the major trends, drivers, investments, vertical players initiatives, government initiatives towards the nanomedicine adoption in the upcoming years along with the details of commercial drugs available in the market. Moreover, the report provides details about the major challenges that are going to impact on the market growth. Additionally, the report gives the complete details about the key business opportunities to key stakeholders to expand their business and capture the revenue in the specific verticals to analyze before investing or expanding the business in this market.

Continued here:
Nanomedicine Market: Clear Understanding of The Competitive Landscape and Key Product Segments - PRnews Leader

MagForce AG: ‘NanoTherm School’ successfully enters the third round with ‘Module B – Part II’ – PharmiWeb.com

DGAP-Media / 20.10.2020 / 09:50

MagForce AG: 'NanoTherm School' successfully enters the third round with 'Module B - Part II'

- MagForce successfully hosted the third session of the practice-oriented, unique and multifaceted application training series for the use of the NanoTherm therapy system for the treatment of glioblastoma.

- The NanoTherm School is part of MagForce's commitment to further optimize the therapy system and train healthcare professionals in its use, and to a broad geographic coverage to increase the availability of the therapy, in order to provide glioblastoma patients with the best care possible.

- The workshop was led by Univ.-Prof. Dr. med. W. Stummer, Director of the Department of Neurosurgery at the University Hospital Mnster

Berlin, October 20, 2020 - With 'Module B - Part II - stereotactic Instillation' MagForce AG (Frankfurt, Scale, Xetra: MF6, ISIN: DE000A0HGQF5), a leading medical device company in the field of nanomedicine focused on oncology, conducted the third round of its NanoTherm School.

The application training series was launched in January 2019 to provide the highest quality of treatment through ongoing support for physicians and certify surgeons in the use of the innovative NanoTherm technology. Same as the prior two sessions which took place in January and November 2019, Module B - Part II focused on advanced techniques in the area of stereotactic instillation and the teaching of the NanoPaste procedure.

Another focus was the exchange of information with the individual treatment centers to provide the best possible care for glioblastoma patients. NanoTherm School is part of MagForce's roll-out strategy, which focuses not only on broad geographical coverage to increase the availability of the therapy, but also on its continuous further optimization and the training of healthcare professionals in the use of the therapy system. The training concept of NanoTherm School was developed in close cooperation with Prof. Dr. Walter Stummer, Dr. Michael Schwake from the University Hospital Mnster and PD Dr. Johannes Wlfer from the Hufeland Klinikum GmbH Mhlhausen, who contribute their many years of experience with the NanoTherm therapy system for the treatment of glioblastoma patients.

Among the workshop participants were also representatives of new clinics that focus on the treatment of brain tumor patients, such as Dr. Georgios Naros, M.D., Senior Physician in the Department of Neurosurgery at the University Hospital of Tbingen and Prof. Dr. Angelika Gutenberg, Head of the Neurosurgery Section at Asklepios Klinikum Hamburg - her feedback: "Excellent, hands-on workshop from MagForce. Organization, motivation and high-end equipment cannot be improved!"

Senior Physician MUDr. Vilem Ruzicka, Paracelsus Klinik Zwickau said: "Thank you for a super organized certification course at the NanoTherm Therapy School of MagForce. It was a very successful and, above all, instructive workshop that provides us as medical practitioners with excellent training in the application of new technologies and therapies. This applies in particular to the very practical training of the various instillation methods and the subsequent evaluation of the results. The preparation and execution were very good, and overall it must be said that the set-up in the 'Medicine in the Green' is excellently suited for such workshops."

About NanoTherm School

Targeted towards medical professionals working in the fields of neuro-surgery and neuro-oncology, the 'NanoTherm Therapy School' series aims at introducing participants to the theoretical knowledge and practical techniques required to successfully apply MagForce's NanoTherm technology for the treatment of glioblastoma. In practicing their skills with human specimens, participants are able to familiarize themselves with the procedures and device usages in a stress-free environment under largely real operating conditions.

The 'NanoTherm School' is designed in three consecutive modules allowing participants to gain knowledge and understanding of MagForce's NanoTherm technology starting from basic application techniques (nanopasting) and building to advanced techniques and new surgical application forms in the final module (Module C). The practical section of the course is complemented by lectures on directly relevant aspects of NanoTherm therapy, as well as sample preparation and surgical videos. The equipment and facilities used during the course meet the requirements of today's state-of-the-art surgical equipment.

For more information about NanoTherm School, please contact:Dipl.-Ing. Tobias Hanitsch (thanitsch@magforce.com) Marcel Pilz (mpilz@magforce.com)

About MagForce AG and MagForce USA, Inc.

MagForce AG, listed in the Scale segment of the Frankfurt Stock Exchange (MF6, ISIN: DE000A0HGQF5), together with its subsidiary MagForce USA, Inc. is a leading medical device company in the field of nanomedicine focused on oncology. The Group's proprietary NanoTherm therapy system enables the targeted treatment of solid tumors through the intratumoral generation of heat via activation of superparamagnetic nanoparticles.

NanoTherm(R), NanoPlan(R), and NanoActivator(R) are components of the therapy and have received EU-wide regulatory approval as medical devices for the treatment of brain tumors. MagForce, NanoTherm, NanoPlan, and NanoActivator are trademarks of MagForce AG in selected countries.

For more information, please visit: http://www.magforce.com.Get to know our Technology: video (You Tube)Stay informed and subscribe to our mailing list

Disclaimer

This release may contain forward-looking statements and information which may be identified by formulations using terms such as "expects", "aims", "anticipates", "intends", "plans", "believes", "seeks", "estimates" or "will". Such forward-looking statements are based on our current expectations and certain assumptions, which may be subject to a variety of risks and uncertainties. The results actually achieved by MagForce AG may substantially differ from these forward-looking statements. MagForce AG assumes no obligation to update these forward-looking statements or to correct them in case of developments, which differ from those, anticipated.

Contact:Barbara von FrankenbergVice PresidentCommunications & Investor RelationsT +49-30-308380-77E-Mail: bfrankenberg@magforce.com

End of Media Release

20.10.2020 Dissemination of a Press Release, transmitted by DGAP - a service of EQS Group AG.The issuer is solely responsible for the content of this announcement.

The DGAP Distribution Services include Regulatory Announcements, Financial/Corporate News and Press Releases. Archive at http://www.dgap.de

More here:
MagForce AG: 'NanoTherm School' successfully enters the third round with 'Module B - Part II' - PharmiWeb.com

Healthcare Nanotechnology (Nanomedicine) Market: Analysis and In-depth Study on Healthcare Nanotechnology (Nanomedicine) Market Size Trends, Emerging…

The Healthcare Nanotechnology (Nanomedicine) market report offers a comprehensive and in-detail assessment of the Healthcare Nanotechnology (Nanomedicine) market and focuses on the key growth contributors of the market to gain a knowledgeable insight on the market. The report contains a detailed account of the history of the Healthcare Nanotechnology (Nanomedicine) market and a thorough and detailed forecast up to the year 2026.

The report takes into account the important factors and aspects that are crucial to the client to post good growth and establish themselves in the Healthcare Nanotechnology (Nanomedicine) market. Aspects such as sales, revenue, market size, mergers, acquisitions, risks, demands, new trends, threats, opportunities, and much more are taken into account to procure a detailed and descriptive research report on the Healthcare Nanotechnology (Nanomedicine) market.

Request Sample Report @ https://beathanreports.com/request-for-sample-report/36794

Description:

This report offers segmented data categorized as per related segments of the Healthcare Nanotechnology (Nanomedicine) market and as the international markets change, the report has documented all the essential aspects that affect the overall growth curve of the market. The reports cover all the segments extensively and offer a detailed explanation of all the factors crucial to growth.

The given report has been assessed to give maximum benefit to our clients and to establish them among the frontrunners in the Healthcare Nanotechnology (Nanomedicine) market. The report has been compiled by using various analyses that have proven to be a game-changer for many in the Healthcare Nanotechnology (Nanomedicine) market. The research sources and tools used by our analysts to assess the report are highly reliable and trustworthy and are approved by industry experts.

The following players are covered in this report:

Amgen

Teva Pharmaceuticals

Abbott

UCB

Roche

Celgene

Sanofi

Merck & Co

Biogen

Stryker

Gilead Sciences

Pfizer

3M Company

Johnson & Johnson

SmitH& Nephew

Leadiant Biosciences

Kyowa Hakko Kirin

Takeda

Ipsen

Endo International

Request Discount About This Report @ https://beathanreports.com/discount-request-on-report/36794

Competitive Analysis:

The report offers effective guidelines and recommendations for players to secure a position of strength and dominance in the Healthcare Nanotechnology (Nanomedicine) market. The report offers extensive coverage of the competition and has a detailed account of the mergers, acquisitions in the Healthcare Nanotechnology (Nanomedicine) market landscape.

The report includes comprehensive data on mergers and acquisitions that will help the clients to get a complete idea of the market competition and also give you extensive knowledge on how to excel ahead and grow in the market.

Breakdown Data by Type

Nanomedicine

Nano Medical Devices

Nano Diagnosis

Other

Healthcare Nanotechnology (Nanomedicine) Breakdown Data by Application

Anticancer

CNS Product

Anti-infective

Other

Based on regional and country-level analysis, the Healthcare Nanotechnology (Nanomedicine) market has been segmented as follows:

North America

United States

Canada

Europe

Germany

France

U.K.

Italy

Russia

Nordic

Rest of Europe

Asia-Pacific

China

Japan

South Korea

Southeast Asia

India

Australia

Rest of Asia-Pacific

Latin America

Mexico

Brazil

Middle East & Africa

Turkey

Saudi Arabia

UAE

Rest of Middle East & Africa

Have Any Query ask to our Expert @ https://beathanreports.com/get-expert-help/36794

Reasons to buy:

See more here:
Healthcare Nanotechnology (Nanomedicine) Market: Analysis and In-depth Study on Healthcare Nanotechnology (Nanomedicine) Market Size Trends, Emerging...

Advanced NanoTherapies Raises $5.3 Million in Seed-Round Financing – Business Wire

LOS GATOS, Calif.--(BUSINESS WIRE)--Advanced NanoTherapies, Inc., a privately-held medical device company committed to exploring nanotechnology for enhanced drug uptake and sustained retention, today announced it has closed on the final tranche of its seed-round financing for a total of $5.3 million as a result of achieving a successful pre-clinical milestone. Funding came from a variety of angel investment sources in the US, Canada, Europe, Asia, and the Middle East, including venture funding from Cleveland Clinic and ForMed Ventures of Taiwan.

Advanced NanoTherapies is led by CEO Marwan Berrada-Sounni, an entrepreneur with extensive experience in percutaneous therapies, including electrophysiology, structural heart, and cardiovascular and endovascular devices.

Our initial focus is on bringing a safer and more effective therapeutic option to patients with peripheral artery disease (PAD) using Sirolimus-coated angioplasty balloons, said Berrada-Sounni. This funding will help us accelerate the development process, build our infrastructure, and move towards bringing this novel technology to patients with PAD.

Advanced NanoTherapies technology platform leverages patented biodegradable functionalized nanoparticles (f-NPs) and a proprietary coating process developed at Cleveland Clinic.

We know that f-NPs enable and improve drug uptake and retention into cells and tissue, said inventor Vinod Labhasetwar, PhD., Professor and Endowed Chair of Nanomedicine, Department of Biomedical Engineering at Cleveland Clinic Lerner Research Institute. Due to technically challenging requirements involved in delivering Sirolimus efficiently to a stenosed artery, Advanced NanoTherapies use of f-NP is an innovative approach.

Advanced NanoTherapies licensed the technology from Cleveland Clinic Innovations.

While drug delivery using coated balloons is a well-established treatment strategy, our vision for using f-NP technology to address PAD represents a major advancement in drug-coated balloon (DCB) technology, said Dr. Mehdi Shishehbor, President, Harrington Heart and Vascular Institute University Hospitals, Cleveland and co-founder and Chief Medical Officer of Advanced NanoTherapies. This technology has implications for many other applications, such as coronary and peripheral in-stent restenosis, below-the-knee tibial disease in CLI, and dialysis AV graft treatment.

Berrada-Sounni added that the company will be working closely with the FDA to achieve an early feasibility study and introduce its technology to human populations.

About Advanced NanoTherapies

Co-founded by Marwan Berrada-Sounni, Advanced NanoTherapies, Inc. is a highly specialized medical device company that is developing a nanoparticle technology-based platform for drug delivery. The company is currently focused on cardiovascular and endovascular applications to bring a safer therapeutic option to patients with PAD using Sirolimus-coated angioplasty balloons. To learn more about Advanced NanoTherapies, visit http://www.AdvNanoT.com.

Go here to read the rest:
Advanced NanoTherapies Raises $5.3 Million in Seed-Round Financing - Business Wire

Deal value in the M&A market decreased in Sept 2020 compared to $35.7 bn in Aug 2020 – Express Pharma

In September 2020, the healthcare industry reported 84 deals worth $28.2 billion as compared to the last 12-month average (September 2019 to August 2020) of 75 deals worth $11.6 billion.

Deal value in the M&A market decreased in September 2020 compared to $35.7 billion in August 2020. Gilead Sciences to acquire Immunomedics, for $88.0 per share in cash, valuing the company at approximately $21 billion contributing 74 per cent of the total deal value during September 2020.

Other notable deals in the month are: Permira, a private equity firm to acquire Neuraxpharm Arzneimittel GmbH, a pharma company focused on the treatment of central nervous system disorders, for $1.8 billion; and Mylan N.V. to acquire the Intellectual Property and Commercialisation Rights of Aspen Pharmacare Holdings Limiteds Thrombosis Business in Europe, for $759 million.

Deal activity in pharma industry in September 2020

VC investments reported a marginal increase in deal value in September 2020

The healthcare industry reported 117 venture capital (VC) deals worth $3.7 billion in September 2020, compared to the last 12-month average (September 2019 to August 2020) of 117 deals worth $2.4 billion.

XtalPi Inc, a computation-driven biotech startup raising $318.8 million in series C round of financing to further develop its ID4 platform, in an attempt to build an AI-powered digital drug R&D infrastructure; Recursion Pharmaceuticals, raising $239.0 million in an oversubscribed series D financing to support the clinical development of its therapeutic pipeline; and InventisBio, a clinical-stage biotech company raising $147 million in Series D financing to support its current products into phase II clinical studies in China and the US, including D-0502 trials in hormone receptor-positive breast cancer and D-0120 trials in gout are the major VC deals reported in September 2020.

Go here to read the rest:
Deal value in the M&A market decreased in Sept 2020 compared to $35.7 bn in Aug 2020 - Express Pharma

Nanorobots Market 2020: Applications, Types and Growing Trends in Market, Gross – News by aeresearch

Business Internet Service Market 2020: Industry Size & Share, Business Strategies, Growth Analysis, Regional Demand By 2025 By Market Study Report

The report published on Business Internet Service Market is an invaluable foundation of insightful data helpful for the decision-makers to form the business strategies related to R&D investment, sales and growth, key trends, technological advan...

The Logistic Software report provides independent information about the Logistic Software industry supported by extensive research on factors such as industry segments size & trends, inhibitors, dynamics, drivers, opportunities & challeng...

The research study 360 Degree Feedback Software market 2020 provides the detailed analysis of the current market status, investment plans, production and consumption, price trends, and analysis by market player, by region, by type, by application e...

The research study APP Store Monetisation market 2020 provides the detailed analysis of the current market status, investment plans, production and consumption, price trends, and analysis by market player, by region, by type, by application etc, an...

The Pumps and Motors report provides independent information about the Pumps and Motors industry supported by extensive research on factors such as industry segments size & trends, inhibitors, dynamics, drivers, opportunities & challenges...

Go here to see the original:
Nanorobots Market 2020: Applications, Types and Growing Trends in Market, Gross - News by aeresearch

Cend Therapeutics Announces Acquisition of Impilo Therapeutics – Yahoo Finance

TipRanks

Is it time for the bears to break out the champagne glasses? Not so fast, says Goldman Sachs. Volatility has ruled the Street for the last few weeks, leading some to conclude that those with a more pessimistic outlook had been vindicated, but the firm believes stocks can still climb higher.According to Goldman Sachs head of U.S. equity strategy, David Kostin, the S&P 500 could still hit 3,600 by the end of the year, and 3,800 by mid-2021, on the back of vaccine-related optimism and progress with the economic reopening. This would reflect gains of 10% and 16%, respectively, should the index ultimately reach these targets.Despite the sharp sell-off in the past week, we remain optimistic about the path of the U.S. equity market in coming months. The Superforecaster probability of a mass-distributed vaccine by Q1 2021 has surged to nearly 70% and economic data show a continuing recovery, Kostin wrote in a recent note. On top of this, the strategist argues the vaccines arrival will push U.S. GDP growth to 6%, compared to the 3.9% consensus estimate.Given Kostins outlook, we wanted to check out three stocks scoring major praise from Goldman Sachs. Not only have they been given a Buy rating, but the firms analysts also see at least 50% upside potential on tap for each. Using TipRanks database, we found out that all three tickers have gotten a thumbs up from analysts at other firms as well. Let's take a closer look.Intellia Therapeutics (NTLA)Focused on utilizing gene editing to develop cell therapies, Intellia Therapeutics wants to stomp out cancer and other immunological diseases for good. Based on its innovative technology, Goldman Sachs recommends that investors pull the trigger.Representing the firm, 5-star analyst Salveen Richter believes that what makes NTLA a stand-out is its use of an adaptive gene editing system based on a proprietary lipid nanoparticle (LNP) delivery method of CRISPR/Cas9 to leverage multiple gene editing strategies. These include the generation of knock-outs (KO) for toxic genes, restoring functional genes by inserting new DNA sequences and the use of consecutive editing combining KO and insertion approaches.We are positive on NTLAs in vivo gene editing approach as it offers a modular system with CRISPR/Cas9 gene editing for functionally curative outcomes. While we note the initial focus is on delivery to the liver, extrahepatic tissue targeting (i.e. CNS) could expand the breadth of NTLAs platform. NTLA is also leveraging its CRISPR/Cas9 editing tools ex vivo to create next-generation engineered cells that can treat oncological and immunological diseases, Richter explained.To this end, the analyst sees several potential catalysts on tap for the next year. Proof-of-concept data for lead program NTLA-2001, its therapy targeting transthyretin amyloidosis (ATTR), a slowly progressive condition characterized by the buildup of abnormal deposits of a protein called amyloid (amyloidosis) in the body's organs and tissues, could come by mid-2021. This data stands to inform the drugs clinical profile (safety/tolerability and early signs of sustained TTR knockdown), which would de-risk NTLAs in vivo editing platform, in Richters opinion.On top of this, IND-enabling studies for NTLA-2002, its therapy designed for hereditary angioedema (HAE), and NTLA-5001, its therapy for WT1+ acute myeloid leukemia (AML), are set to kick off in 2021. Richter estimates that peak sales for both candidates could reach $895 million and $806 million, respectively, with data from both also validating the breadth of editing approaches (knockouts and/or insertions).If that wasnt enough, Richter cites the ongoing NVS-led Phase 1/2 OTQ923 sickle cell disease (SCD) trial as a possible upside driver. While we note the limited economics to NTLA from this program and competitor dynamics with bluebird bios (BLUE) LentiGlobin and CRISPR Therapeutics (CRSP) CTX001 that are ahead in clinical development, the study should serve as proof-of-concept for the platform. First data could be presented in 2021, the analyst commented.All of this prompted Richer to initiate coverage with a Buy rating and $33 price target. This target conveys her confidence in NTLAs ability to climb 50% higher in the next year. (To watch Richters track record, click here)Looking at the consensus breakdown, 3 Buys and 2 Holds have been published in the last three months. Therefore, NTLA gets a Moderate Buy consensus rating. Based on the $37.13 average price target, shares could rise 67% in the next year. (See NTLA stock analysis on TipRanks)Vir Biotechnology (VIR)Moving on to another healthcare company, Vir Biotechnology is developing a broad portfolio of product candidates that are designed to combat serious, global infectious diseases in new ways. With it standing at the front of the pack in the COVID-19 monoclonal antibody (mAb) race, its no wonder Goldman Sachs likes what its seeing.Firm analyst Paul Choi cites a recent data readout from one of VIRs competitors as reaffirming his confidence. On September 16, Eli Lilly reported interim data from the Phase 2 BLAZE-1 trial evaluating its mAb therapies, LY-CoV555 and LY-CoV016, in mild or moderate COVID-19 patients. The data revealed that treatment with LY-CoV555 led to a roughly 72% reduction in the need for hospitalization, with no safety signals observed.Choi also points out that the results were more pronounced in high risk patients (age or BMI) as most study hospitalizations across both groups occurred in patients with these underlying risk factors.While resistant viral variants did appear in 8% of LY-CoV555-treated patients and 6% of patients on placebo, management has stated that competing single or multiple mAb cocktail approaches might not be optimized, with viral escape mutants potentially emerging. VIR argues its approach is differentiated given the high barrier to resistance, potent effector function, potential for increased lung tissue concentration and extended half-life.Even though VIR is behind its peers in terms of development timelines, Choi thinks that the company is making substantial progress. VIR recently initiated the Phase 2/3 COMET-ICE study of VIR-7831, its mAb for COVID-19, as a monotherapy (versus a combination approach) in patients with mild or moderate COVID-19. Initial data is set to be released by the end of 2020, with top-line data expected in January. Weighing in on the above, Choi commented, In the absence of preclinical binding affinity data from LY-CoV555, it is premature to hypothesize on the potential for VIR-7831 to demonstrate improved efficacy vs. the competing antibodies; however, we see the LLY data as establishing proof-of-concept for antibodies in COVID-19 while also setting an attainable bar for future antibody monotherapy/cocktail treatments. Moreover, we view the addressable market for COVID-19 antibodies as significant enough to support several approved therapies in the indication in the near-term.In line with his optimistic approach, Choi reiterated his Buy rating and $54 price target. Should the 5-star analysts thesis play out, a twelve-month gain of 69% could potentially be in the cards. (To watch Chois track record, click here)Is the rest of the Street in agreement? The majority of other analysts are. 4 Buys, 1 Hold and 1 Sell have been issued in the last three months, so the word on the Street is that VIR is a Moderate Buy. With the average price target clocking in at $51.67, shares could jump 61% in the next year. (See VIR stock analysis on TipRanks)Peloton Interactive (PTON)Switching gears now, we move on to Peloton Interactive. The company, which offers exercise bikes and remote workout classes, rose to fame at the start of the COVID-19 pandemic. After its fiscal Q4 earnings results blew estimates out of the water, Goldman Sachs believes this stock has more room to run.In the most recent quarter, PTON posted revenue of $607.1 million, beating the $586.2 million consensus estimate and reflecting a 172% year-over-year increase. This is up from growth of 65.6% in the previous quarter. Adjusted EBITDA came in at $143.6 million, ahead of the Streets $73.5 million call. Management pointed to heightened demand during the COVID-19 crisis and significantly lower marketing spend as the drivers of this strong showing.Goldman Sachs Heath Terry tells clients he was especially excited about the Connected Fitness segments performance. Connected Fitness product revenue landed at $486 million, up 199% year-over-year, while customer deposits and deferred revenue grew 300% year-over-year. The five-star analyst also highlights the fact that subscriber net adds were 205,000, versus 174,100 net adds in fiscal Q3 2020 and guidance of 154-164,000.As for PTONs forward-looking guidance, Terry was also impressed. While the company guided fiscal Q1 2021 and FY21 revenue and adjusted EBITDA well above consensus, given the backlog of demand exiting the June quarter and the 6-8 weeks of deliveries already on order by consumers, we expect this guidance will again prove overly conservative, he explained.This performance prompted Terry to state, We continue to believe that Peloton represents a significant long-term opportunity as the company is in the earliest stages of creating new and expanding existing categories of connected fitness products, an opportunity that we believe has been permanently accelerated by the current COVID-19 crisis.It should be noted that the company faces significant risks going forward. These include new entrants, evolving consumer tastes as well as execution challenges. That being said, Terrys bullish thesis remains very much intact.Expounding on this, the analyst said, ... we believe that the window of opportunity for any meaningful competitor is rapidly closing, something that, along with the large and expanding addressable market for Pelotons high ARPU, high margin, extremely low churn subscription business, remains underappreciated by the market, even with the stocks recent outperformance.It should come as no surprise, then, that Terry stayed with the bulls. To this end, he kept a Buy rating and $138 price target on the stock. Investors could be pocketing a gain of 53%, should this target be met in the twelve months ahead. (To watch Terrys track record, click here)In general, other analysts are on the same page. PTONs Strong Buy consensus rating breaks down into 20 Buys, 2 Holds and 1 Sell. The $112.05 average price target brings the upside potential to 23%. (See PTON stock analysis on TipRanks)To find good ideas for stocks trading at attractive valuations, visit TipRanks Best Stocks to Buy, a newly launched tool that unites all of TipRanks equity insights.Disclaimer: The opinions expressed in this article are solely those of the featured analysts. The content is intended to be used for informational purposes only. It is very important to do your own analysis before making any investment.

Originally posted here:
Cend Therapeutics Announces Acquisition of Impilo Therapeutics - Yahoo Finance