Category Archives: Stem Cell Medicine

Global NK Cell Therapy and Stem Cell Therapy Market Analysis, Growth, Size, Demand & Forecast 2020-2025 – Cole of Duty

The latest trending report Global NK Cell Therapy and Stem Cell Therapy Industry Market to 2025 available at MarketStudyReport.com is an informative study covering the market with detailed analysis. The report will assist reader with better understanding and decision making.

The research report on NK Cell Therapy and Stem Cell Therapy Industry market report comprises of an in-depth analysis of this industry vertical. The key trends that describe the NK Cell Therapy and Stem Cell Therapy Industry market during the forecast period are cited in the document, alongside additional factors including industry policies and regional scope. Moreover, the study specifies the impact of prevailing industry trends on potential investors.

Request a sample Report of NK Cell Therapy and Stem Cell Therapy Industry Market at:https://www.marketstudyreport.com/request-a-sample/2707408?utm_source=algosonline.com&utm_medium=PSR

COVID-19, the disease it causes, surfaced in late 2020, and now had become a full-blown crisis worldwide. Over fifty key countries had declared a national emergency to combat coronavirus. With cases spreading, and the epicentre of the outbreak shifting to Europe, North America, India and Latin America, life in these regions has been upended the way it had been in Asia earlier in the developing crisis. As the coronavirus pandemic has worsened, the entertainment industry has been upended along with most every other facet of life. As experts work toward a better understanding, the world shudders in fear of the unknown, a worry that has rocked global financial markets, leading to daily volatility in the U.S. stock markets.

The report also provides with an overview of the competitive landscape along with a thorough analysis of the raw materials as well as the downstream buyers.

Revealing a summary of the competitive analysis of NK Cell Therapy and Stem Cell Therapy Industry market:

An overview of the regional scope of the NK Cell Therapy and Stem Cell Therapy Industry market:

Ask for Discount on NK Cell Therapy and Stem Cell Therapy Industry Market Report at:https://www.marketstudyreport.com/check-for-discount/2707408?utm_source=algosonline.com&utm_medium=PSR

Other takeaways from the NK Cell Therapy and Stem Cell Therapy Industry market report:

Significant Features that are under Offering and Key Highlights of the Reports:

Key questions answered in the report:

For More Details On this Report: https://www.marketstudyreport.com/reports/covid-19-outbreak-global-nk-cell-therapy-and-stem-cell-therapy-industry-market-report-development-trends-threats-opportunities-and-competitive-landscape-in-2020

Some of the Major Highlights of TOC covers:

Development Trend of Analysis of NK Cell Therapy and Stem Cell Therapy Market Market

Marketing Channel

Market Dynamics

Methodology/Research Approach

Related Reports:

1. Global Stress Tests Equipments Market Research Report 2015-2027 of Major Types, Applications and Competitive Vendors in Top Regions and CountriesStress Tests Equipments market research report provides the newest industry data and industry future trends, allowing you to identify the products and end users driving Revenue growth and profitability. The industry report lists the leading competitors and provides the insights strategic industry Analysis of the key factors influencing the market.Read More: https://www.marketstudyreport.com/reports/global-stress-tests-equipments-market-research-report-2015-2027-of-major-types-applications-and-competitive-vendors-in-top-regions-and-countries

2. Global Cardiac Resynchronization Therapy Device Market Research Report 2015-2027 of Major Types, Applications and Competitive Vendors in Top Regions and CountriesCardiac Resynchronization Therapy Device Market report characterize imperative Portion and contenders of the market regarding market estimate, volume, esteem. This report likewise covers every one of the locales and nations of the world, which demonstrates a territorial improvement status, it additionally incorporates Business Profile, Introduction, Revenue and so on.Read More: https://www.marketstudyreport.com/reports/global-cardiac-resynchronization-therapy-device-market-research-report-2015-2027-of-major-types-applications-and-competitive-vendors-in-top-regions-and-countries

Read More Reports On: https://www.marketwatch.com/press-release/at-168-cagr-screen-fingerprint-sensor-market-size-set-to-register-50244-million-usd-by-2025-2020-06-12

Contact Us:Corporate Sales,Market Study Report LLCPhone: 1-302-273-0910Toll Free: 1-866-764-2150 Email: [emailprotected]

Read this article:
Global NK Cell Therapy and Stem Cell Therapy Market Analysis, Growth, Size, Demand & Forecast 2020-2025 - Cole of Duty

Calquence showed long-term efficacy and tolerability for patients with chronic lymphocytic leukaemia in two trials | Vaccines | News Channels -…

DetailsCategory: VaccinesPublished on Saturday, 13 June 2020 12:40Hits: 358

ACE-CL-001 trial showed an overall response rate of 97% with a sustained safety profile for previously untreated patients after more than four years

In pivotal ASCEND trial, 82% of patients with relapsed or refractory disease treated with Calquence remained progression free at 18 months vs. 48% for comparators

LONDON, UK I June 12, 2020 I Detailed results from both the Phase II ACE-CL-001 trial and the pivotal Phase III ASCEND trial showed the long-term efficacy and tolerability of Calquence (acalabrutinib) in chronic lymphocytic leukaemia (CLL), one of the most common types of adult leukaemia.1,2,3

The results will be presented during the Virtual Edition of the 25th European Hematology Association (EHA) Annual Congress, 11 to 14 June 2020.

In the single-arm ACE-CL-001 trial, 86% of CLL patients treated with Calquence as a 1st-line monotherapy remained on treatment at a median follow up of more than four years. The trial showed an overall response rate of 97% (7% complete response; 90% partial response) and a 100% overall response rate in subgroups of patients with high-risk disease characteristics, including genomic aberrations (17p deletion and TP53 mutation), immunoglobulin mutation status (unmutated IGHV), and complex karyotype. Safety findings showed no new long-term issues.1,4

In the final analysis of ASCEND, an estimated 82% of patients with relapsed or refractory CLL treated with Calquence remained alive and free from disease progression at 18 months compared with 48% of patients on rituximab combined with idelalisib or bendamustine.2 The trial previously met the primary endpoint of Independent Review Committee-assessed progression-free survival at the interim analysis.5

Richard R. Furman, Director of the CLL Research Center, Weill Cornell Medicine said: These data demonstrate no new safety concerns for acalabrutinib, confirming its ability to safely provide meaningful, long-term clinical benefit for patients with treatment-naive and relapsed or refractory disease. The safety profile of acalabrutinib makes treatment to progression an important and plausible option for patients.

Jos Baselga, Executive Vice President, Oncology R&D said: These long-term data reaffirm that Calquence delivers a durable response with a favourable safety profile for chronic lymphocytic leukaemia patients. Patients with chronic lymphocytic leukaemia are typically 70 years or older with comorbidities and often require treatment over a long time, making the sustained safety and efficacy profile highly relevant to their quality of life.

Results from the Phase II ACE-CL-001 trial informed the development of the pivotal Phase III ELEVATE TN trial, which, along with findings from the Phase III ASCEND trial, formed the basis for the US approval of Calquence for the treatment of patients with CLL or small lymphocytic lymphoma (SLL).

Calquence in previously untreated CLL: 4.4-year follow-up from Phase II trial (abstract #S163)

The Phase II ACE-CL-001 trial investigated safety and efficacy of Calquence (100mg twice-daily [n=62] or 200mg once-daily [n=37]) in previously untreated patients with CLL.1 On 1 May 2015, patients receiving the 200mg dosing regimen were switched to the 100mg regimen.1

Key data from the Calquence Phase II ACE-CL-001 trial1

CI, confidence interval; CR, complete response; DoR, duration of response; EFS, event free survival; TTR, time to response; NR, not reached; ORR, overall response rate; PR, partial response

Response rates were 100% in each subgroup of patients with high-risk disease characteristics (unmutated IGHV [n=57], 17p deletion [n=9], TP53 mutation [n=9], and complex karyotype [n=12]), and reduction in lymph node disease was noted in all patients tested (n=97).1

At the time of data cut-off, 85 (86%) patients receiving Calquence remained on treatment. Six patients discontinued treatment due to adverse events (AEs) and three patients discontinued for progressive disease (PD). No patient discontinued Calquence due to bleeding events, hypertension, or atrial fibrillation. Incidence of AEs generally diminished with time on the trial. The most common AEs (40%) of any grade in the trial were diarrhoea (52%), headache (45%), upper respiratory tract infection (44%), arthralgia (42%), and contusion (42%). All-grade and Grade 3 events of clinical interest included infection (84% and 15%, respectively), bleeding events (66%, 3%), hypertension (22%, 11%), leukopenia (9%, 9%), and thrombocytopenia (3%, 1%). Atrial fibrillation (all grades) occurred in 5% of patients with Grade 3 occurring in 2%. Second primary malignancies (SPM) excluding non-melanoma skin (all grades) occurred in 11% of patients.1 Serious adverse events (SAEs) were reported in 38% of patients. SAEs occurring in more than two patients included pneumonia (n=4) and sepsis (n=3).1

Final results of Calquence Phase III ASCEND trial in relapsed or refractory CLL (abstract #S159)

ASCEND was a global, randomised, multicentre, open-label, Phase III trial that investigated the efficacy and safety of Calquence (100mg twice-daily) versus investigators choice of rituximab combined with idelalisib (IdR) or bendamustine (BR) in patients with relapsed or refractory CLL.2

Key data from the final analysis of the Calquence Phase III ASCEND trial2

BR, rituximab in combination with bendamustine; CI, confidence interval, DoR, duration of response; HR, hazard ratio; IdR, rituximab in combination with idelalisib; INV, investigator; NR, not reached; ORR, overall response rate; OS, overall survival; PFS, progression-free survival

Sixteen per cent of patients on Calquence, 56% of patients on IdR, and 17% of patients on BR discontinued treatment because of AEs. Common AEs occurring in greater than 15% of patients of any grade in the Calquence arm of the trial included headache (22%), neutropenia (21%), diarrhoea (20%), upper respiratory tract infection (20%), cough (16%), and anaemia (16%). Events of clinical interest for Calquence versus controls included atrial fibrillation (all grade, 6% and 3%, respectively), major haemorrhage (all grade, 3% in both arms), infections (Grade 3, 20% and 25%, respectively), and SPM excluding non-melanoma skin cancer (all grade, 5% and 2%, respectively). SAEs (any grade) occurred in 33% of patients receiving Calquence, 56% of IdR patients, and 26% of BR patients.2

Chronic lymphocytic leukaemia

Chronic lymphocytic leukaemia (CLL) is one of the most common types of leukaemia in adults, with an estimated 105,000 new cases globally in 2016 and 21,040 new cases in the US in 2020, and the number of people living with CLL is expected to grow with improved treatment as patients live longer with the disease.3,6,7,8 In CLL, too many blood stem cells in the bone marrow become abnormal lymphocytes and these abnormal cells have difficulty fighting infections.3 As the number of abnormal cells grows there is less room for healthy white blood cells, red blood cells, and platelets.3 This could result in anaemia, infection, and bleeding.3 B-cell receptor signalling through Brutons tyrosine kinase is one of the essential growth pathways for CLL.

Calquence

Calquence(acalabrutinib) is a next-generation, selective inhibitor of Brutons tyrosine kinase (BTK).Calquencebinds covalently to BTK, thereby inhibiting its activity.4,9 In B-cells, BTK signaling results in activation of pathways necessary for B-cell proliferation, trafficking, chemotaxis, and adhesion.4

Calquenceis approved for the treatment of adult patients with chronic lymphocytic leukaemia (CLL) in nine countries and for adult patients with mantle cell lymphoma (MCL) who have received at least one prior therapy in 14 countries. The US MCL indication is approved under accelerated approval based on overall response rate. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials. As part of an extensive clinical development programme, AstraZeneca and Acerta Pharma are currently evaluatingCalquencein 23 company-sponsored clinical trials.Calquenceis being developed for the treatment of multiple B-cell blood cancers including CLL, MCL, diffuse large B-cell lymphoma, Waldenstrm macroglobulinaemia, follicular lymphoma, and other haematologic malignancies.

AstraZeneca in haematology

Leveraging its strength in oncology, AstraZeneca has established haematology as one of four key oncology disease areas of focus. The Companys haematology franchise includes two US FDA-approved medicines and a robust global development programme for a broad portfolio of potential blood cancer treatments. Acerta Pharma serves as AstraZenecas haematology research and development arm. AstraZeneca partners with like-minded science-led companies to advance the discovery and development of therapies to address unmet need.

AstraZeneca in oncology

AstraZeneca has a deep-rooted heritage in oncology and offers a quickly growing portfolio of new medicines that has the potential to transform patients' lives and the Company's future. With six new medicines launched between 2014 and 2020, and a broad pipeline of small molecules and biologics in development, the Company is committed to advance oncology as a key growth driver for AstraZeneca focused on lung, ovarian, breast and blood cancers. In addition to AstraZeneca's main capabilities, the Company is actively pursuing innovative partnerships and investments that accelerate the delivery of our strategy, as illustrated by the investment in Acerta Pharma in haematology.

By harnessing the power of four scientific platforms - Immuno-Oncology, Tumour Drivers and Resistance, DNA Damage Response and Antibody Drug Conjugates - and by championing the development of personalised combinations, AstraZeneca has the vision to redefine cancer treatment and one day eliminate cancer as a cause of death.

AstraZeneca

AstraZeneca (LSE/STO/NYSE: AZN) is a global, science-led biopharmaceutical company that focuses on the discovery, development and commercialisation of prescription medicines, primarily for the treatment of diseases in three therapy areas - Oncology, Cardiovascular, Renal and Metabolism, and Respiratory & Immunology. Based in Cambridge, UK, AstraZeneca operates in over 100 countries and its innovative medicines are used by millions of patients worldwide. Please visitastrazeneca.comand follow the Company on Twitter@AstraZeneca.

Media

For details on how to contact the Investor Relations Team, please click here. For Media contacts, click here.

References

1. Byrd JC, et al. Acalabrutinib in Treatment-Nave Chronic Lymphocytic Leukemia: Mature Results From Phase 2 Study Demonstrating Durable Remissions and Long-Term Tolerability. Abstract S163 presented at the Virtual Edition of the 15th European Hematology Association (EHA) Annual Meeting. Available online. Accessed June 2020.

2. Ghia P, et al. Acalabrutinib (Acala) vs Idelalisib plus Rituximab (IdR) or Bendamustine plus Rituximab (BR) in Relapsed/Refractory (R/R) Chronic Lymphocytic Leukemia (CLL): ASCEND Final Results. Abstract S159 presented at the Virtual Edition of the 15th European Hematology Association (EHA) Annual Meeting. Available online. Accessed June 2020.

3. National Cancer Institute. Chronic Lymphocytic Leukemia Treatment (PDQ)Patient Version. Available online. Accessed June 2020.

4.Calquence(acalabrutinib) [prescribing information]. Wilmington, DE; AstraZeneca Pharmaceuticals LP; 2019.

5. Ghia P, et al. ASCEND Phase 3 Study of Acalabrutinib vs Investigators Choice of Rituximab Plus Idelalisib (IdR) or Bendamustine (BR) in Patients with Relapsed/Refractory (R/R) Chronic Lymphocytic Leukemia (CLL). Abstract LB2606 at the 2019 European Hematology Association (EHA) Annual Meeting. Available online. Accessed June 2020.

6. Global Burden of Disease Cancer Collaboration. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2016. JAMA Oncol. 2018;4(11):1553-1568.

7. American Cancer Society. Key Statistics for Chronic Lymphocytic Leukemia. Available online. Accessed June 2020.

8. Jain N, et al. Prevalence and Economic Burden of Chronic Lymphocytic Leukemia (CLL) in the Era of Oral Targeted Therapies. Blood. 2015;126:871.

9. Wu J, Zhang M & Liu D. Acalabrutinib (ACP-196): a selective second-generation BTK inhibitor.J Hematol Oncol. 2016;9(21).

SOURCE: AstraZeneca

Read the rest here:
Calquence showed long-term efficacy and tolerability for patients with chronic lymphocytic leukaemia in two trials | Vaccines | News Channels -...

High-throughput intracellular biopsy of microRNAs for dissecting the temporal dynamics of cellular heterogeneity – Science Advances

Abstract

The capability to analyze small RNAs responsible for post-transcriptional regulation of genes expression is essential for characterizing cellular phenotypes. Here, we describe an intracellular biopsy technique (inCell-Biopsy) for fast, multiplexed, and highly sensitive profiling of microRNAs (miRNAs). The technique uses an array of diamond nanoneedles that are functionalized with size-dependent RNA binding proteins, working as fishing rods to directly pull miRNAs out of cytoplasm while keeping the cells alive, thus enabling quasi-single-cell miRNA analysis. Each nanoneedle works as a reaction chamber for parallel in situ amplification, visualization, and quantification of miRNAs as low as femtomolar, which is sufficient to detect miRNAs of a single-copy intracellular abundance with specificity to single-nucleotide variation. Using inCell-Biopsy, we analyze the temporal miRNA transcriptome over the differentiation of embryonic stem cells (ESCs). The combinatorial miRNA expression patterns derived by inCell-Biopsy identify emerging cell subpopulations differentiated from ESCs and reveal the dynamic evolution of cellular heterogeneity.

Embryonic stem cells (ESCs) are self-renewable and can differentiate into all types of cells in an adult organism. They are increasingly used in disease modeling, drug discovery, and regenerative medicine (13). More recently, the emergence of induced pluripotent stem cells (iPSCs) eludes the potential of patient-specific regeneration of damaged or diseased tissues (4, 5) and brings stem cellbased therapy to the forefront of the development of previously unidentified treatments for many diseases that are challenging for traditional methods. One of the issues for these efforts is the development of protocols to ensure directed differentiation of stem cells both in vitro and in vivo (6), which relies on the understanding and controlling of the cellular heterogeneity generated from stem cell differentiation and is critical for the clinical adoption of relevant therapeutic strategies (7).

MicroRNAs (miRNAs), a class of noncoding small RNAs, were reported to be involved in the regulation of self-renewal and differentiation of many stem cells (8). They regulate gene expression by binding to specific mRNA targets to promote mRNA degradation or translational inhibition. Many recent studies have shown a close relationship between miRNA transcriptome and cellular heterogeneity in different tissues (9, 10) or over stem cell differentiation (11, 12). However, in situ profiling of miRNAs in living cells is still challenging, hindering the adoption of miRNA as a therapeutic indicator in clinical practices (13). The trace amount of miRNAs in cytoplasm requires a profiling technique to be highly specific and sensitive (13, 14). A size selection step is typically required, following the isolation of total RNA from cellular homogenate (13, 15). Because of the short length of miRNAs, small primers are required in polymerase chain reaction (PCR) and often cause reduced priming efficiency, nonspecific hybridization, and, thus, erroneous results (14).

Among the tools for miRNA profiling, quantitative reverse transcription PCR (qRT-PCR) is the gold standard, but it is rather a validation instead of discovery tool. Deep sequencing resolves miRNA transcriptome with an extremely high throughput, but it is, however, disadvantaged by high cost, long turnover time, and complex data analysis (13). Both methods inherit similar issues from the required PCR process as mentioned above. Alternatively, microarray provides multiplexed miRNA analysis based on probe-target hybridization but has lower specificity and sensitivity (13, 14). All these commonly used techniques require isolation of RNA sample from cellular lysates, providing only average measurements of miRNAs for all cells. Consequently, important information about the heterogeneity of the cell populations would be missing and is only accessible by using single cellbased analytical techniques (16). Some in situ miRNA assay tools were recently reported by combining high-resolution microscopy with nanotechnology (17, 18), which are less quantitative and sometimes limited by toxicity issues.

Here, we describe an intracellular biopsy (inCell-Biopsy) technique for multiplexed in situ profiling of miRNAs in living cells. An array of diamond nanoneedles were functionalized with RNA binding protein (p19) and were used as fishing rods to directly pull multiple targeted miRNAs out of cell cytoplasm in a few minutes, leaving the cells alive. After the inCell-Biopsy operation, each nanoneedle then worked as a separated reaction plant for parallel in situ amplification, visualization, and quantification of miRNAs. The detection limit can reach as low as 1015 M, which is almost three orders of magnitude lower than the abundance of a single copy in a cell. Using inCell-Biopsy, we demonstrated multiplexed profiling of miRNAs in living cells and analyzed the temporal miRNA transcriptome over the differentiation of ESCs toward motor neurons, revealing the cellular heterogeneity and associated evolutionary dynamics of the differentiated cell populations based on miRNA expression.

The inCell-Biopsy technique is based on the continuous development of a molecular fishing system (19), which uses an array of diamond nanoneedles as fishing rods for minimum-invasive and reversible access of cytoplasmic regions of mammalian cells (Fig. 1A and movie S1). Specifically, for fishing miRNAs, a size-dependent RNA binding protein, p19, was cross-linked to functionalize the nanoneedles, working as the fishing hook to capture double-strand RNAs (dsRNAs) (Fig. 1A; more details in fig. S1). P19 can selectively bind to all dsRNAs of 20 to 22 base pairs (bp) (20, 21), which is a range covering almost all miRNAs in mammalian cells. As mature intracellular miRNAs are mostly single stranded (22), in this study, for each targeted miRNA, a bait RNA sequence was delivered to the cytoplasm to hybridize with the targets for p19 to capture. When the functionalized diamond nanoneedles are interfaced with live cells using a centrifugation facilitated procedure, the fishing rods can penetrate the cell membrane to access the cytoplasmic region via a temporary membrane disruption, which simultaneously facilitates the intracellular delivery of the bait RNAs (fig. S1) (19, 23). Upon the retrieval of the nanoneedles, the targeted miRNAs are isolated, leaving the cells alive (viability, 96.2 1.5%; means SD, n = 3).

(A) Schematic illustration of intracellular biopsy of miRNAs from live cells. (B) On-needle amplification of miRNA signals by hybridization chain reaction (HCR). (C) DNase-assisted multiple rounds of signal visualization. (D) Image processing and informatic approaches for miRNA transcriptome analysis.

After the inCell-Biopsy operation, a hybridization chain reaction (HCR) was performed on the nanoneedles to amplify the miRNAs. The HCR is featured by two single-strand DNAs (ssDNAs) with a stem-loop structure (hairpin 1 and 2), which can cyclically hybridize with each other if the stem part of one sequence is open (Fig. 1B) (24). For each miRNA target, the corresponding bait RNA contains the complementary sequence plus a small encoding overhang part at its 3 end, which binds to a small DNA sequence (initiator) to trigger the HCR (Fig. 1B). To enhance the detection sensitivity, one of the hairpins (hairpin 1) was fluorescently labeled and was quenched until its stem opening in the HCR. For multiplexing, the overhang part of the bait sequence was uniquely encoded for different miRNA targets, so that multiple miRNA targets can be visualized by different fluorophores (table S1). While the number of optically separable fluorophores may be limited (e.g., four channels), we implemented multiple rounds of HCR by removing the DNA hairpins after collecting the signals at the end of each round using deoxyribonuclease I (DNase I) enzyme, so that a new set of miRNAs could be examined to improve the analytical throughput of the inCell-Biopsy technique (Fig. 1C). Three rounds of HCR enabled us to examine 12 miRNAs after each biopsy. To enhance assay reliability, we dedicated one of the four channels to a reference miRNA (Fig. 1D), cel-miR-39, which does not exist in human or rodent cells and was artificially introduced to the cell cytoplasm, thus to eliminate potential systemic errors caused by experimental variations. The expression level of a targeted miRNA was indicated by the normalized fluorescence (with respect to the reference) acquired by confocal microscopy. While the nanoneedles could not be correlated to each cell with one-to-one mapping, the scattered signal from thousands of nanoneedles still retains the rich information about the cell population based on their miRNA expression (Fig. 1D).

To characterize the detection limit of our inCell-Biopsy technique, we first performed a mock experiment by profiling miRNAs from medium containing a premixed dsRNA (miR-34a and corresponding bait sequence) at different concentrations varying from 1016 to 1010 M. A nanoneedle chip was incubated in the solution and then rinsed before proceeding to further analysis. For every chip, signals from more than 1000 nanoneedles were collected and quantified (Fig. 2, A and B). Our results showed reliable differentiation of dsRNA of tested concentrations down to 1015 M (Fig. 2C). The overall profile of the fluorescence intensities was observed to positively associate with the miRNA concentrations (Fig. 2D), and the ratio of positive nanoneedles also exhibited an association with the miRNA concentration, roughly following the Langmuir isotherm model (Fig. 2E; also see note S1) (25). To test the detection specificity, we performed an assay to detect the synthetic let-7a miRNA over sequences with 1nucleotide (nt) mismatch (let-7c) or 2-nt mismatches (let-7b), and showed that the inCell-Biopsy technique is specific to single-nucleotide variation by successfully discriminating closely related miRNA sequences (Fig. 2F). The inCell-Biopsy was then implemented to detect miRNAs (let-7a or miR-34a) in cultured A549 cells (Fig. 2G). The two miRNAs were reported to express with different abundance in the cells: Let-7a is highly expressed, and miR-34a is of relatively low intracellular level (26). The nanoneedle chip was interfaced with A549 cells using a centrifugation-controlled method to initiate the intracellular biopsy (19, 23) and to deliver the two bait sequences to the intracellular domain. After a 15-min fishing reaction, the chip was retrieved from cells for analysis. For successful miRNA biopsy, the nanoneedles were identified to colocalize with the fluorescence signals from HCR amplification (Fig. 2H), which is significantly higher than different controls (Fig. 2, I and J; also see fig. S2A). The HCR amplification is especially useful in the detection of miRNAs of low intracellular abundance (e.g., miR-34a), which would otherwise be unobservable without the on-needle HCR amplification (fig. S2B).

(A) SEM image of the diamond nanoneedles; scale bar, 50 m. (B) Fluorescence images (top view) showing miRNA signals (red) on the nanoneedles (green); scale bar, 50 m. For (A) and (B), the boxed region is enlarged below; scale bars, 10 m. (C) Analysis of detection limit with violin plots showing the distribution of miRNA signals from all nanoneedles. *P < 0.005 by Kruskal-Wallis test. a.u., arbitrary units. (D) Relationship between miRNA concentration and fluorescence averaged from all nanoneedles. The red line indicates logarithmic fit (R2 = 0.99, P < 0.001 by F test). (E) Relationship between miRNA concentration and ratio of signal+ nanoneedles. The blue dashed line indicates a nonlinear fit by Langmuir isotherm model (R2 = 0.98, P < 0.001 in F test). (F) Analysis of detection specificity. NC, no target included. *P < 0.001 by ANOVA test. (G) Image of A549 cells after treatment; scale bar, 50 m. (H) Fluorescence visualization of miRNAs (red, let-7a or miR-34a) on the nanoneedles (green); scale bars, 10 m for three-dimensional and top view, 1 m for enlarged view. (I) Comparison of miRNA (let-7a or miR-34a) signals from different controls. (J) Ratio of signal+ nanoneedles for experiments with or without HCR amplification. For (D), (E), (I), and (J), n = 3; the error bar indicates SEM; *P < 0.001 by ANOVA test.

The capability to capture the dynamics of miRNA expression is extremely important for a profiling technique, as intracellular miRNAs play a key regulatory role in gene expression networks and change when cells switch their status (27). We then applied inCell-Biopsy to characterize relevant miRNAs in cells undergoing DNA damage or at different stages of a cell cycle to show its potential as a technique for probing cellular dynamics. Upon ultraviolet-induced DNA damage, let-7a was significantly down-regulated, and miR-16 and miR-26 were significantly up-regulated within just several minutes (fig. S3). In an extended temporal window as the cells progress to different division cycles, let-7a, miR-21, and miR-34a were observed to gradually increase from G1, to S, to G2 stage, while miR-24 remained stable at these stages (fig. S4). These results echo well with the literature (28, 29) and demonstrated inCell-Biopsys capability to monitor the fluctuation of miRNAs expression associated with cellular activity. In addition, the technique not only provides an overall assessment of miRNA expression in the cells but also captures the dynamic heterogeneity of cell populations, as confirmed by flow cytometry (fig. S4D) and qRT-PCR analysis (fig. S4, F and G).

After the above technical validations, we next applied inCell-Biopsy to investigate the temporal miRNA transcriptome and its relationship to cellular heterogeneity over the differentiation of mouse ESCs (mESCs) (HB9: GFP) toward motor neurons. We chose to profile nine different miRNAs in these cells at day 0, day 7, and day 14 from the induction of differentiation (Fig. 3, A and B). By using a custom-developed processing streamline and program (fig. S5), the expression of the nine miRNAs at various differentiation stages were obtained (Fig. 3C). When all the data were pooled together blindly, t-distributed stochastic neighbor embedding (t-SNE) (30) quantification showed the overall evolutionary change of the cells by the appearance of three self-organized clusters along with ESC differentiation, suggesting the validity of using combinatory miRNA expression pattern to indicate cell identity in this process (Fig. 3D).

(A) Experimental design of monitoring miRNA dynamics over the differentiation of embryonic stem cells (ESCs). RA, retinoic acid; and SAG, smoothened agonist. (B) Phase (Ph)contrast and fluorescence (green fluorescence indicates GFP) images showing morphological change of the cells along with differentiation. Scale bars, 50 m. (C) Confocal fluorescence image (top view) of diamond nanoneedles after fishing and HCR amplification. Scale bar, 20 m. The boxed region is enlarged below to show the expression of nine miRNAs from three rounds of amplification and visualization. Scale bars, 2 m. (D) t-SNE clustering of the pooled multidimensional miRNA vectors that resulted from inCell-biopsy at all three stages, showing the overall evolution of miRNA expression along with ESC differentiation.

Cells generated from ESC differentiation are typically heterogeneous (31), the inCell-Biopsy technique provides us the possibility to decipher this heterogeneity and biogenic evolution by using temporal miRNA dynamics (16). For each of the nine miRNA targets, we quantified its fold change at later differentiation stages with respect to the initial stem cell level, performed a self-diffusionbased spectral clustering (details in Materials and Methods Section) for the multidimensional miRNA measurements from thousands of nanoneedles pooled from six independent replicates, and determined the optimal numbers of clusters by eigengap (fig. S6A) (32). It was found that stable subpopulations were clearly observed at both 7 and 14 days after differentiation, and the cells appeared to be more scattered at the later stage (Fig. 4, A and B). The clustering results were also confirmed by t-SNE analysis (Fig. 4C) as well as principle component analysis (fig. S6B). The heatmaps and violin plots of the nine miRNAs showed the unique expression pattern of each cluster at a specific differentiation stage (Fig. 4, D, E, G, and H and fig. S7) and also suggested some similarities of particular clusters across different stages (e.g., cluster 3 at day 7 versus cluster 5 at day 14), implicating potential evolutionary correlation between them. Statistically, we were able to identify cluster-specific miRNA expression signatures (table S2). The shared signature between clusters of day 7 and day 14 supports our speculation of their evolutionary relationship. For instance, cluster 3 of day 7 and cluster 5 of day 14 shared the similar signature miRNAs of miR99a, miR218, and miR9. miR24, miR218, and miR219 are the shared signature miRNAs of cluster 4 of day 7 and cluster 3 of day 14. The discovery of these clusters was made by statistical analysis of signals from thousands of nanoneedles that were interfaced with a large population of cells. The proportion of a particular cluster (out of all nanoneedles) was also evaluated as an indicator for the percentage of a cell subpopulation, assuming an even distribution of the nanoneedles on a uniform culture of cells (Fig. 4F). This scattering information would be lost if averaged miRNA measurements were performed with cell lysate as what are mostly done by existing methods.

Self-diffusionbased spectral clustering and associated similarity network for the multidimensional miRNA measurements from thousands of nanoneedles at day 7 (A) or day 14 (B) of differentiation. (C) Separation of cellular subpopulations indicated by t-SNE analysis at day 7 or day 14. Heatmap showing distinct miRNA expression patterns between different clusters obtained from unsupervised classification at day 7 (D) or day 14 (E). (F) Sector graph showing the proportion of nanoneedles in each cluster out of the total number of nanoneedles on day 7 or day 14. Radar plots (left) and associated violin plots (right) show the averaged expression of the nine miRNAs for each of the identified clusters at day 7 (G) or day 14 (H).

To study the evolutionary correlation among different cell subpopulations (represented by the clusters) over differentiation, we used the multidimensional miRNA data from inCell-Biopsy to study the statistical association between the clusters of day 7 and day 14 to determine the closest pairs. Each cluster of day 14 can be uniquely traced back to link with a cluster of day 7 (P < 0.001, hypergeometric tests; Fig. 5A). For example, cluster 3/4/5 of day 14 was respectively found to be most correlated with cluster 4/2/3 of day 7; these paired clusters also showed similar miRNA expression patterns as shown in the violin plots (Fig. 4, G and H). Clusters 1 and 2 of day 14 both traced back to cluster 1 of day 7, suggesting cluster 2 of day 14 to be a newly differentiated subpopulation derived from cluster 1 of day 7 (Fig. 5B).

(A) Hypergeometric tests for determining the closest pair of clusters from the two differentiation stages. Significant associations are labeled by red squares, colored in proportion to log10(P value) (all P < 0.001). (B) The phylogenetic tree shows the evolutionary relationship among the clusters (cell subpopulations) as differentiation proceeds. The widths of the branches are proportionate to transformed P values [log10(P values)] derived from hypergeometric tests. (C) Correlation between the cluster miRNA pattern (derived from inCell-biopsy) with the results acquired by miR-seq for sorted motor neurons/progenitors. *P < 0.001. (D) Quantitative analysis of the averaged expression of the nine miRNAs for motor neuronlike clusters at day 7 and day 14. Error bars indicate SEM from six independent experiments. (E) Density histograms and associated violin plots showing the variation and distribution of nine miRNAs expression for motor neuronlike clusters at day 7 and day 14.

To figure out the relationship between the identified cluster based on miRNA expression and the cell type identity, we then focused on differentiated motor neuron/progenitors (GFP+) and compared the inCell-Biopsyacquired miRNA profile for each cluster to the data acquired by miRNA sequencing (miR-seq) for sorted motor neuron/progenitors at various differentiation stages. We found that the majority of the nine-dimensional miRNA vectors in cluster 3 of day 7 or cluster 5 of day 14 showed significantly higher correlation to the miR-seqmeasured miRNA expression pattern (P < 0.001, Wilcoxon signed-rank test; Fig. 5C), which was not observed for the other clusters of the same differentiation stage, suggesting that the nanoneedles in cluster 3 of day 7 or cluster 5 of day 14 mostly sampled motor neurons/progenitors in the miRNA biopsy operation. Taking a closer look at the two clusters, we further observed dynamic changes of different miRNAs (Fig. 5, D and E). For example, miR-294, a stem cellspecific miRNA (33), was significantly reduced from day 7 to day 14, whereas the motor neuronenriching miR-9 and miR-218 (34) were significantly increased over the same period (Fig. 5D). In addition, for the two motor neuron-like clusters, the miRNA expression was generally more scattered at day 14 (compared with day 7; Fig. 5E), which suggested an increased variation of the cell status as differentiation progresses.

Here, we develop a highly versatile and powerful technique, inCell-Biopsy, for in situmultiplexed profiling of miRNAs in living cells. The technique is capable of cherry picking targeted miRNAs from cell cytoplasm while leaving the samples intact afterward. For quantitative analysis of cellular RNAs, most of the existing techniques (e.g., qRT-PCR, microarray, RNA sequencing) started with RNA samples extracted from a population of cells and only provide an averaged measurement of the cell population (13, 14). InCell-Biopsy, on the other hand, isolates targeted miRNAs from a large number of individual cells within just a few minutes by using a diamond nanoneedlefacilitated molecular fishing system (19), and parallels in situ amplification, visualization, and quantification of miRNAs using each nanoneedle as a separated reaction chamber. In this way, our method not only detects averaged miRNA expression level but also captures the cellular heterogeneity of a cell population based on miRNA profiling, which is typically missed in other methods and only accessible using single-cell RNA sequencing (scRNA-seq) analysis (16). In this study, the density of the diamond nanoneedles was roughly controlled at ~5 nanoneedles per 10 by 10 m2 region. Although we cannot establish an exact one-to-one (or multiple-to-one) contacting map between the nanoneedles and each individual cell, the inCell-Biopsy technique enables a quasisingle-cell analysis to provide rich information for characterizing cell mixtures by using multidimensional miRNA profiles. As a proof of concept, we used the inCell-Biopsy technique to dissect cellular heterogeneity over the differentiation of ESCs and investigated the evolution of the cells with a dynamic temporal miRNA transcriptome analysis.

While the intracellular biopsy strategy has been previously reported (19, 3537), in this study, such a concept was further elaborated with specific biochemical design targeting multiple miRNAs, along with a complete framework for multiplexed in situ signal amplification, visualization, and quantification, which altogether are formulated as a quasisingle-cell miRNA profiling platform. Notably, the diamond nanoneedles are rigid enough to puncture cell membrane and remain ultraelastic at nanoscale to sustain the deformation without fracture during an inCell-biopsy operation (38). Although different nanostructures were recently developed as tools to isolate intracellular materials from living cells (3537) and may have the potential to be used for detecting miRNAs when combined with sequencing techniques, our inCell-biopsy technique stands out with a balanced combination of in situ capability, high throughput, ease of use, and independence of expensive equipment.

As one of the core merits, the inCell-Biopsy does not involve any cell lysis and RNA preparation procedures; therefore, the examined cells can be preserved for further longitudinal analysis. This feature also markedly simplifies the experimental operation, reduces the processing time, and provides the opportunity to quantitatively examine the temporal dynamics of miRNA expression for the same batch of cells receiving external stimuli or undergoing internal switch of cellular programs. It would be extremely useful when miRNA profiling is used as a characterization or quality control for cell-based therapeutic treatment (39). Meanwhile, the capability to directly fish miRNAs from the cytoplasm of an individual cell effectively bypasses the dilution of low-abundance miRNAs and prevents sample loss during cell lysing and RNA extraction procedures. Although only a single copy of miRNA is presented in a cell, the actual concentration for nanoneedle-assisted inCell-Biopsy would be around 1013 to 1012 M, which is well tolerated by the detection limit of the technique (1015 M).

Our inCell-Biopsy is based on an intracellular molecular fishing system, in which diamond nanoneedles are used as the fishing rod and RNA binding protein (p19) is used as the fishing hook, which specifically binds to dsRNA (not to ssRNA or dsDNA) in a size-dependent manner (20). For an inCell-Biopsy operation, the dsRNA complexes are formed by the hybridization between the targeted single-strand miRNA and a complementary bait sequence, which was thought to diffuse into cell cytosol via the nanopuncture-induced reversible membrane disruption (23). It is also possible that cytosolic materials could diffuse to cells outside, but this should not be an issue for the treated cells in this study, as the nanoneedles were tightly interfaced with the mobile lipid bilayer membrane (fig. S1), making it less likely for intracellular components (e.g., miRNAs) to leak out and be captured. Notably, intracellular pri-miRNAs or pre-miRNAs that lead to false positive in traditional PCR-based detections (13) would not interfere with our assay, because their structures would prohibit bait hybridization and subsequent binding to p19 proteins. The introduction of an encoded bait sequence for each miRNA target further enhances the specificity of the inCell-Biopsy technique. Practically, p19 can bind to all available dsRNAs of the right length, so that multiplexed detection (e.g., nine miRNAs in this study) can be easily implemented by effortless intracellular delivery of multiple bait sequences (20). Compared with scRNA-seq, the throughput of the inCell-Biopsy technique may be lower at the current stage, but it has, undoubtedly, advantages in substantially lower cost and more efficient experimental protocol. For improvement, the fluorescence labeling system can also be fine-tuned to include more channels to improve the assay throughput. For example, if the hairpin sequences were labeled with quantum dots, it would be easy to achieve an eight-channel imaging system, and three rounds of imaging would increase the throughput to 21 miRNA targets. In addition, the incorporation of certain barcoding strategy (e.g., nanostring system) (40) can further increase the analytical throughput to allow the analysis of hundreds of miRNA targets within a single visualization cycle.

As we had demonstrated, a nine-dimensional miRNA vector space produced by inCell-Biopsy already carries rich information for the identification of heterogeneous clusters that represent the cellular subpopulations differentiated from ESCs. The clustering was autonomously derived from a quasisingle-cell analysis of the miRNA expression patterns, which have recently been reported to be a good indicator for cellular heterogeneity (11, 12). While we cannot spatially determine the contacting relationship of each nanoneedle to an individual cell, statistically, the miRNA profiling by inCell-Biopsy can still reflect the compositional nature of examined cells, assuming a random but uniform distribution of the diamond nanoneedles over an evenly cultured cell. The multidimensional miRNA vector derived from each nanoneedle is treated as an input to a huge miRNA vector space, which effectively created a quasisingle-cell analysis framework for miRNA transcriptome analysis. Particularly, in this study, the correspondence between nanoneedle clusters and identity of specific cell subpopulation was verified to further confirm the validity of the analytical results acquired by inCell-Biopsy. For example, we used a protocol to direct motor neuron differentiation and blindly identified a major nanoneedle cluster (out of all nanoneedles) that was highly correlated to motor neuron identity at both day 7 and day after differentiation. However, when the same mESCs (HB9: GFP) were undergoing spontaneous differentiation without the induction compounds (retinoic acid and smoothened agonist), the motor neuron-like cluster was not discoverable (fig. S8).

The capability to retain cell sample after inCell-Biopsy operation enables multiround miRNA profiling at different time points, thus providing a temporal miRNA transcriptome analysis. Our results show that the inCell-Biopsy not only creates a quick snapshot of the heterogeneity of the examined live cells based on their miRNA profiles but also captures the temporal dynamics of miRNA expression, and it subsequently gives the cellular evolutionary path, as well as the biogenic relationship among heterogeneous emerging cell populations, which is especially informative for clinical applications (7).

In summary, we demonstrate a novel and powerful technique, inCell-Biopsy, for profiling miRNAs in living cells. The temporal miRNA dynamics captured by this technique can be used to reveal the evolution of cellular heterogeneity in mixed cell populations over extended culture periods, potentially providing a quick and convenient evaluation platform for the quality control of the emerging therapeutic strategies involving cell components.

HB9: GFP mESCs were acquired from the Stem Cell Core Facility of Columbia University. ESCs were seeded in a petri dish coated with 0.1% gelatin and were further cultured in an incubator at 37C with 5% CO2 for proliferation. After 3 days, ESCs were trypsinized for cell seeding. Typically, 250 ml of ESC culture medium consisted of 200 ml of EmbryoMax Dulbecco's modified Eagle's medium (DMEM) (Millipore), 37.5 ml of fetal bovine serum (FBS; Hyclone), 2.5 ml of EmbryoMax MEM Nonessential Amino Acids (Millipore), 2.5 ml of nucleosides (Millipore), 2.5 ml of 200 mM l-glutamine (Invitrogen), 2.5 ml of penicillin/streptomycin (pen/strep) (10,000 U/ml penicillin and 10,000 g/ml streptomycin, Invitrogen), 180 l of diluted 2-mercaptoethanol [diluted 1:100 in phosphate-buffered saline (PBS) with Mg and Ca; Invitrogen], and 25 l of leukemia inhibitory factor (LIF)/ESGRO (Millipore). Afterward, the embryonic stem medium was replaced by differentiation medium. Typically, 450 ml of differentiation medium consisted of 200 ml of Advanced DMEM/F12 (Invitrogen), 200 ml of Neurobasal Medium (Invitrogen), 46 ml of KnockOut Serum Replacement (Invitrogen), 4.6 ml of pen/strep, 4.6 ml of l-glutamine, and 320 l of diluted 2-mercaptoethanol. After 2 days, retinoic acid (RA; diluted 1:1000 in differentiation medium; Sigma-Aldrich) and smoothened agonist (SAG; dilutes 1:1000 in differentiation medium; Sigma-Aldrich) were added into the medium for a strong induction to motor neuron differentiation. After 3 days of in vitro differentiation, the differentiation medium supplemented with 4.5 l of glial-derived neurotrophic factor (Invitrogen), 9 ml of B27 (Invitrogen), and 4.5 ml of N2 supplement (Invitrogen) was used for a better motor neuron growth.

A549 cancer cells were maintained in DMEM (Life Technologies) supplemented with 10% FBS, l-glutamine, and pen/strep. Before molecular fishing experiments, cells were seeded in a four-well multidish (Nunclon) and allowed to grow until ~80% confluence.

The fabrication of diamond nanoneedles follows a protocol as previously described (19), involving the deposition of nanodiamond film and subsequent bias-assisted reactive ion etching (RIE) by electron cyclotron resonance microwave plasma chemical vapor deposition (ECR-MPCVD). N-type (001) silicon wafers of 3-inch diameter were used as a substrate. Before nanodiamond deposition, the substrate was soaked in ultrasonic bath for 60 min in ethanol, containing a suspension of nanodiamond powders with a grain size of 5 nm. Nanodiamond films 7 m thick were then deposited using a commercial ASTeX MPCVD equipped with a 1.5-kW microwave generator. The nanodiamond deposition was performed in the plasma induced in 10% CH4/H2 mixture at a total pressure of 30 torr and total gas flow rate of 200 standard cubic centimeter per minute (SCCM). The microwave power and deposition temperature were maintained at 1200 W and 800C, respectively. After the nanodiamond film deposition, RIE was performed using ECR-MPCVD. The ASTeX microwave source used a magnetic field of 875 G generated by an external magnetic coil. The RIE used a mixture of 45% Ar and 55% H2 as the reactive gases, which were supplied at a total flow rate of 20 SCCM. The substrate bias was 200 V, and the reactant pressure was 7 103 torr. The etching duration was 3 hours, and the input microwave power was 800 W. The morphology of the resulted diamond nanoneedles was characterized by scanning electron microscopy (SEM; Philips FEG SEM XL30), and the sample was tilted 90 for SEM.

To functionalize the diamond nanoneedles with p19 protein, a patch was first bathed in piranha (3:1, v/v; 98% H2SO4:27.5% H2O2) solution at 90C for 1.5 hours and then cleaned by distilled water, methanol, methanol/dichloride methane (DCM) mixture (3:1, v/v), and DCM sequentially. The nanoneedle patch was dried with nitrogen and then immersed in (3-aminopropyl)triethoxysilane solution (20% in DCM, v/v) overnight in a nitrogen-protected environment. Ethanol, isopropyl alcohol, and distilled water were sequentially used to rinse the nanoneedle patch, which would be further dried by nitrogen blow. The nanoneedle patch was then bathed in NHS-biotin solution (1 g/ml in PBS; Sigma-Aldrich) for 1 hour, streptavidin (20% of the streptavidin was labeled by fluorescent dye, Alexa Fluor 488) solution (10 g/ml in PBS; Invitrogen) for 2 hours, and biotinylated p19 siRNA binding protein solution (1 g/ml in PBS; New England Biolabs) for 1 hour. The nanoneedle patch was rinsed with distilled water between adjacent bath steps. After each experiment, the nanoneedle patch was soaked in hot (~90C) piranha solution (3:1, v/v; 98% H2SO4:27.5% H2O2) to remove all cross-linked materials (protein, nucleotides, etc.) on the nanoneedle surface, and SEM images were taken ensure the integrity of the nanoneedle structure. In this way, one patch can be used for at least 20 times. All the materials were acquired from Sigma-Aldrich unless otherwise specified.

Intracellular delivery of RNA bait sequences and miRNA fishing were performed using a centrifugation-controlled process (19). For cells in adherent culture, the medium was first removed, and 100 l of RNA bait sequence solution (10 nM for every bait sequence in serum-free medium) was applied to the cells. A nanoneedle patch was then placed facing toward the cells in a four-well petri dish. The whole complex was then placed in a centrifuge with a plate rotor and spun at 400 revolutions per minute (rpm) (22.8 g) for 4 min. After first centrifugation, the setup was placed in an incubator for 10 min to allow the bait sequences to diffuse into cytoplasm and to form dsRNA with their intracellular targets. Afterward, a second centrifugation was performed to enhance miRNA fishing results. During a centrifugation, the ramping speed was controlled to maintain a smooth acceleration to avoid any movement of the nanoneedle patch on cells. For acceleration and deceleration, 3 and 6 rpm/s were, respectively, selected.

Each RNA bait sequence used in the inCell-Biopsy has a unique 10-bp overhang sequence that can be used to amplify the signal coupling with the HCR (41). To perform the on-needle HCR amplification of miRNAs, the initiator sequences were diluted in the hybridization buffer containing 5 sodium citrate (SSC; Invitrogen) with 0.05% tween (pH 7.4; Invitrogen) to a final concentration of 10 nM; the hairpins were diluted in reaction buffer to a final work concentration of 20 nM. After a rinse with 0.05% SDS for 15 min, the nanoneedle patch was immersed in the initiator solution for hybridization between the dsRNAs and the initiator sequences. Following a quick rinse with the wash buffer (1 SSC, 0.05% tween), the patch was further incubated at 37C for 3 hours in hybridization buffer containing different hairpin DNAs (20 nM) with FAM, JOE, CY3, or CY5 fluorescent labels and black hole quencher. After the initiation of the HCR, the heterodimer was separated, and the absorption/emission of the fluorophore was restored. To guarantee the separation and reduce the requenching effect after the binding of hairpins 1 to 2, a short unmatched sequence was included in the hairpin sequences to work as a spacer. All hairpin and initiator oligonucleotides were acquired from BGI (Shenzhen, China) and summarized in table S1.

After first imaging, the HCR-amplified nanoneedle patch was immersed into DNase I solution for 1 hour in 37C to fully elute the DNA hybridized on the needle surface, followed by washing three times with wash buffer. After that, the nanoneedle patch was enabled to perform the HCR amplification to detect another four miRNA targets.

After each HCR amplification process, confocal microscopy (Leica SP8, 40 objective with 1.3 numerical aperture, water immersion) was performed to visualize and quantify the miRNAs captured on the surface of diamond nanoneedles. A nanoneedle patch was scanned with 0.3-m z resolution to get a stack of 45 to 55 slices, and both three-dimensional reconstruction and maximal projection of the stack were acquired. As a result of the HCR amplification, the fluorescent speckles (from DNA hairpin sequences) on nanoneedle surface was quantified and used to analyze the fishing of miRNA targets from the cells inside. After three rounds of amplification, images were projected and aligned to get a 12-channel image stack containing the fluorescence intensity and relative position information. To differentiate the positive signals from background noises or debris, fluorescent speckles of 0.4 to 1.5 m in diameter were firstly selected as nanoneedle regions, and a fluorescence threshold was then applied to sort out the positive nanoneedles with captured miRNAs. For each nanoneedle patch, we lastly obtained a matrix of intensities representing miRNA expression levels, where the columns are miRNA targets and the rows are different nanoneedles.

After obtaining the miRNA expression matrix, we divided the expression data of day 7 and day 14 by matched average value of that at embryo stem cell stage followed by log2 transformation to derive miRNA expression fold change data. To make sure that the miRNA expression data are comparable between different replicates, quantile normalization was subsequently performed for day 7 and day 14, respectively.

Having performed the preprocessing, the miRNA expression fold change data from all the replicates were combined as input for unsupervised classification at day 7 and day 14, respectively. We performed self-diffusion analysis (42), which was implemented by propagating the affinity matrices to improve sample similarities learning, followed by spectral clustering (32) for its relatively better adaptability to data distribution and the lower time consumption. We calculated eigengap (32) based on local scaling affinity, which infers the self-tuning affinity of sample-by-sample distances, and chose the optimal number (k) of clusters for clustering (fig. S6A). The sample-by-sample similarity matrices and similarity networks for day 7 and day 14 are shown in Fig. 4 (A and B), respectively.

For each miRNA profiling at day 7 (or day 14), we calculated Pearson correlation coefficients (PCCs) between its miRNA expression and the average expression levels of nanoneedles in different clusters at day 14 (or day 7). The nanoneedle at day 7 was subsequently assigned to the most correlated cluster at day 14 and vice versa. A confusion matrix was constructed to summarize the total number of nanoneedles classified simultaneously to each pair of clusters at day 7 and day 14, and a hypergeometric test was subsequently performed to evaluate the statistical significance of their association. First, we calculated the average expression profile of probes in each cluster of day 7. Second, for each nanoneedle in day 14, we calculated PCC with the average profile of each cluster of day 7, and the nanoneedle (day 14) was paired to the cluster (day 7) with the highest PCC. Third, all paired day 14day 7 nanoneedle relationships were counted, summarized, and followed by a hypergeometric test for overrepresentation of nanoneedles in day 14 paired to a cluster in day 7. Last, P values derived from the hypergeometric tests were adjusted for multiple hypothesis testing using the Benjamini-Hochberg procedure and illustrated as a heatmap. Similarly, using clusters of day 14 as a reference, we did association tests for the nanoneedles of day 7, and the conclusion is highly consistent.

To illustrate the potential evolutionary relationship between clusters at different stages, a phylogenetic tree was generated on the basis of the statistical associations between clusters at day 7 and day 14, where the branch width represents the transformed P value [log10(P)] derived from the hypergeometric tests. For validation, motor neuron cells (GFP+) were isolated with Sony SH800S cell sorter; the total RNA of sorted cells was extracted using the TRIzol reagent kit (Life Technologies) for miR-seq analysis (BGI). To investigate the potential cell type identities of nanoneedle clusters, we calculated PCCs between the inCell-Biopsyacquired miRNA profiles and the expression levels of the nine miRNAs measured by miR-seq.

To measure miRNAs (let-7a, miR-21, miR-24, miR-34a, and RNU43) using qRT-PCR, the total RNA was extracted from A549 cells using the TRIzol reagent kit (Life Technologies). For 15-l reactions, 10 ng of total RNA was reverse-transcribed and analyzed by the TaqMan miRNA Assays kit (product no. 4366596; Life Technologies). The expression of a particular miRNA was analyzed using the Applied Biosystems real-time PCR instrument following the manufacturers protocol.

At least three independent biological replicates were used for all experiments (n 3); for each replicate, signals from at least 250 nanoneedles were collected for analysis. For Fig. 2C, the bin size along the y axis was 60 (fluorescence, arbitrary units) for the violin plots; the whisker range of the overlaying boxplots is 1 to 99%, and each box shows 25, 50, and 75% percentile of the data. Kruskal-Wallis analysis was performed to determine the statistical significance among different dsRNA concentrations, and P < 0.005 indicates a significant difference. For Fig. 2 (H and I), analysis of variance (ANOVA) was performed to determine the statistical significance; P < 0.05 indicates a significant difference. The error bars indicate SEM from three independent experiments. For Fig. 4 (G and H), the bin size along the y axis was 0.1 (fold change of the expression level) for the violin plots. To identify cluster-specific miRNA expression signatures, a two-tailed Students t test was performed to assess whether each miRNA is differentially expressed between a specific cluster and the other clusters of day 7 (or day 14). For each cluster of day 7 (or day 14), the miRNA expression signatures were prioritized on the basis of the absolute log2 expression level (|log2EL| > = 0.75) and Benjamini-Hochbergadjusted P value (P < 0.001). For Fig. 5C, Wilcoxon signed-rank test was performed to determine the statistical significance; P < 0.001 indicates a significant difference. For Fig. 5D, the error bars indicate SEM from six independent experiments. For Fig. 5E, the bin size along the y axis was 0.1 (fold change of the expression level) for the violin plots.

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license, which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.

Acknowledgments: Funding: This work was supported by the National Natural Science Foundation of China (81871452, 81802384, and 51772318), by the Science, Technology and Innovation Committee of Shenzhen Municipality (JCYJ20170818100342392, JCYJ20180507181624871, and JCYJ20170413141236903), by the General Research Fund (11278616, 11203017, 11102317, 11103718, and 11103619) from the Research Grants Council of Hong Kong SAR, and by the Health and Medical Research Fund (06172336) from the Food and Health Bureau of Hong Kong SAR. Author contributions: P.S. conceived the project, designed, and supervised the research. Z.W., X.Z., and K.X. carried out the experiments and analyzed the data. L.Q. and X.W. performed the statistical and bioinformatics analysis. Y.Y. and W.Z. provided the diamond nanoneedle array. M.L., E.H.C.C., X.J., and L.H. helped with the experiments. All authors contributed to the writing of the manuscript. Competing interests: P.S., W.Z., L.H., X.W., and Z.W. are inventors on a pending patent related to this work (no. US15/875,385, filed 18 January 2018). The authors declare that they have no competing interests. Data materials and availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

Read the original post:
High-throughput intracellular biopsy of microRNAs for dissecting the temporal dynamics of cellular heterogeneity - Science Advances

BrainStorm to Present at the Raymond James Human Health Innovations Conference – PRNewswire

NEW YORK, June 11, 2020 /PRNewswire/ --BrainStorm Cell Therapeutics Inc.(NASDAQ: BCLI), a leading developer of adult stem cell therapies for neurodegenerative diseases, today announced Chaim Lebovits, CEO and Ralph Kern, MD, MHSc, President and Chief Medical Officer, will present a corporate overview on Thursday, June 18 at 9:00 am EST, during theRaymond James Human Health Innovations Conference, a virtual event connecting institutional investors with company management teams that will be held June 15-18, 2020.

Mr. Lebovits and Dr. Kern will update conference participants on the Company's investigational therapeutic, NurOwn, that is currently in a fully enrolled phase 3 study for the treatment of ALS and a phase 2 study for the treatment of progressive multiple sclerosis. Additionally, they will present an overview of the Company's financial position and pipeline. After the presentation, the management team will participate in a question and answer session with institutional investors.

Mr. Lebovits and Dr. Kern will be joined by David Setboun, PhD, MBA, Chief Operating Officer, Stacy Lindborg, PhD, Head of Global Clinical Research, and Preetam Shah, PhD, MBA, Chief Financial Officer, for a series of one-on-one meetings, with select institutional investors arranged by Raymond James.

Participants can view the presentation via the event link and those unable to join will have access to an archived link on the Company's Events and Presentation webpage after the conclusion of the conference.

EVENT: Raymond James Human Health Innovations Conference

PRESENTATION: Thursday, June 18th at 9:00 am EST

LINK: https://bit.ly/2YmZf8u

About NurOwn

NurOwn (autologous MSC-NTF) cells represent a promising investigational therapeutic approach to targeting disease pathways important in neurodegenerative disorders. MSC-NTF cells are produced from autologous, bone marrow-derived mesenchymal stem cells (MSCs) that have been expanded and differentiated ex vivo. MSCs are converted into MSC-NTF cells by growing them under patented conditions that induce the cells to secrete high levels of neurotrophic factors. Autologous MSC-NTF cells can effectively deliver multiple NTFs and immunomodulatory cytokines directly to the site of damage to elicit a desired biological effect and ultimately slow or stabilize disease progression. BrainStorm has fully enrolled a Phase 3 pivotal trial of autologous MSC-NTF cells for the treatment of amyotrophic lateral sclerosis (ALS). BrainStorm also recently receivedU.S.FDA acceptance to initiate a Phase 2 open-label multicenter trial in progressive MS and enrollment began inMarch 2019.

AboutBrainStorm Cell Therapeutics Inc.

BrainStorm Cell Therapeutics Inc.is a leading developer of innovative autologous adult stem cell therapeutics for debilitating neurodegenerative diseases. The Company holds the rights to clinical development and commercialization of the NurOwn technology platform used to produce autologous MSC-NTF cells through an exclusive, worldwide licensing agreement. Autologous MSC-NTF cells have received Orphan Drug status designation from theU.S. Food and Drug Administration(U.S.FDA) and theEuropean Medicines Agency(EMA) in ALS. BrainStorm has fully enrolled a Phase 3 pivotal trial in ALS (NCT03280056), investigating repeat-administration of autologous MSC-NTF cells at sixU.S.sites supported by a grant from theCalifornia Institute for Regenerative Medicine(CIRM CLIN2-0989). The pivotal study is intended to support a filing forU.S.FDA approval of autologous MSC-NTF cells in ALS. BrainStorm also recently receivedU.S.FDA clearance to initiate a Phase 2 open-label multicenter trial in progressive Multiple Sclerosis. The Phase 2 study of autologous MSC-NTF cells in patients with progressive MS (NCT03799718) started enrollment inMarch 2019.

Safe-Harbor Statement

Statements in this announcement other than historical data and information, including statements regarding future clinical trial enrollment and data, constitute "forward-looking statements" and involve risks and uncertainties that could causeBrainStorm Cell Therapeutics Inc.'sactual results to differ materially from those stated or implied by such forward-looking statements. Terms and phrases such as "may", "should", "would", "could", "will", "expect", "likely", "believe", "plan", "estimate", "predict", "potential", and similar terms and phrases are intended to identify these forward-looking statements. The potential risks and uncertainties include, without limitation, BrainStorm's need to raise additional capital, BrainStorm's ability to continue as a going concern, regulatory approval of BrainStorm's NurOwn treatment candidate, the success of BrainStorm's product development programs and research, regulatory and personnel issues, development of a global market for our services, the ability to secure and maintain research institutions to conduct our clinical trials, the ability to generate significant revenue, the ability of BrainStorm's NurOwn treatment candidate to achieve broad acceptance as a treatment option for ALS or other neurodegenerative diseases, BrainStorm's ability to manufacture and commercialize the NurOwn treatment candidate, obtaining patents that provide meaningful protection, competition and market developments, BrainStorm's ability to protect our intellectual property from infringement by third parties, heath reform legislation, demand for our services, currency exchange rates and product liability claims and litigation,; and other factors detailed in BrainStorm's annual report on Form 10-K and quarterly reports on Form 10-Q available athttp://www.sec.gov. These factors should be considered carefully, and readers should not place undue reliance on BrainStorm's forward-looking statements. The forward-looking statements contained in this press release are based on the beliefs, expectations and opinions of management as of the date of this press release. We do not assume any obligation to update forward-looking statements to reflect actual results or assumptions if circumstances or management's beliefs, expectations or opinions should change, unless otherwise required by law. Although we believe that the expectations reflected in the forward-looking statements are reasonable, we cannot guarantee future results, levels of activity, performance or achievements.

CONTACTS

Investor Relations:Preetam Shah, MBA, PhDChief Financial OfficerBrainStorm Cell Therapeutics Inc.Phone: +1-862-397-1860[emailprotected]

Media:

Sean LeousWestwicke/ICR PRPhone: +1-646-677-1839[emailprotected]

SOURCE Brainstorm Cell Therapeutics Inc

More here:
BrainStorm to Present at the Raymond James Human Health Innovations Conference - PRNewswire

Stem Cell Assay Market Report 2020 by Global Key Players, Types, Applications, Countries, Market Size, Forecast to 2026 (Based on 2020 COVID-19…

Stem Cell Assay market report 2020 entitles with an in-depth analysis towards the competitive market, which involves the market shares and company outline of the major competitors functioning in the Stem Cell Assay market. The study offers detailed summarization of products, various technologies applied in the Stem Cell Assay type of product, and manufacturing analysis taking in to account all the major factors that include cost, revenue, gross profit and so on. This Stem Cell Assay report consists of a financial overview, market synopsis, demand towards various segments and growth aspects. Numerous applications, and analysis on demand and supply activities, Stem Cell Assay market price during the projected period. The global Stem Cell Assay market report will be maintaining good productivity with increasing CAGR of XX%. Considering all the basic aspects such as product type, Stem Cell Assay application, various industrial competitors, and regional analysis.

Request for a sample report here https://www.orbisresearch.com/contacts/request-sample/4718175

Prominent players included in the global Stem Cell Assay market:

Bio-Techne CorporationBio-Rad LaboratoriesPromega CorporationMerck KGaAHemogenixGE HealthcareCellular Dynamics InternationalSTEMCELL TechnologiesCell BiolabsThermo Fisher Scientific

The global Stem Cell Assay market division by product type:

Viability/CytotoxicityIsolation & PurificationCell IdentificationProliferationDifferentiationFunctionApoptosis

The global Stem Cell Assay market division by application:

Regenerative Medicine & Therapy DevelopmentDrug Discovery and DevelopmentClinical Research

Market bifurcation by Stem Cell Assay geographical region includes North America, Europe, Latin America, Middle East and Africa, Asia-Pacific, and Rest of the world. respectively.

The Stem Cell Assay report represents an important tool towards the manufacturers all over the world along with the value chain and also for new competitors by allowing them to benefit from business opportunities as well advanced business tactics. It has studied the Stem Cell Assay market thoroughly focusing on market size, growth opportunities, and market status.

Detailed qualitative Stem Cell Assay research approach which involves investigation and recognition of the following terms:

* Stem Cell Assay market classification

* Driving factors influencing Stem Cell Assay growth

* Stem Cell Assay key restraints and market opportunities

* Upcoming product developments and Stem Cell Assay major challenges

* SWOT (Strength, Weakness, Opportunities, and Threat) and Stem Cell Assay PORTERS Five Forces analysis

The Stem Cell Assay market report serves major analytics on the market position of the Stem Cell Assay industries and acts a valuable source for guiding and directing the companies interested in the Stem Cell Assay market as well individuals. The Stem Cell Assay market report explains the basic fundamentals related to the market strategies their applications, product specifications, definitions, various classifications, production process, their cost structure, Stem Cell Assay raw materials analysis and even more.

Ask our Expert if You Have a Query at: https://www.orbisresearch.com/contacts/enquiry-before-buying/4718175

Questions answered in the global Stem Cell Assay market report:

1. What are the market strategies applicable, market insight, and Stem Cell Assay product type analysis?

2. What are driving factors influencing the growth of the global Stem Cell Assay market, analysis by region and application?

3. What are the market dynamics, that involves the scope of the product and price breakdown of Stem Cell Assay key manufacturers?

4. Who are the major challenges, opportunities and risk factors for Stem Cell Assay market, including the upstream and downstream towards raw material and buyers?

5. Who are the key market players, Stem Cell Assay business outline by application, product type, market share and gross profit?

6. What are major threats tackled by the sellers in the global Stem Cell Assay market?

The Stem Cell Assay current and past data related to market signifies the existing market valuation and the future prospects. Moreover, data collected here are through primary and secondary research, that includes interviews with major Stem Cell Assay industries including the values of top manufacturers, their suppliers, and various application, as well company report, latest trends, and reviews. Also, through different research findings, Stem Cell Assay distribution channels, traders, results, and Appendix.

About Us:

Orbis Research (orbisresearch.com) is a single point aid for all your market research requirements. We have vast database of reports from the leading publishers and authors across the globe. We specialize in delivering customized reports as per the requirements of our clients. We have complete information about our publishers and hence are sure about the accuracy of the industries and verticals of their specialization. This helps our clients to map their needs and we produce the perfect required market research study for our clients.

Contact Us:

Hector CostelloSenior Manager Client Engagements4144N Central Expressway,Suite 600, Dallas,Texas 75204, U.S.A.Phone No.: USA: +1 (972)-362-8199 | IND: +91 895 659 5155Email ID: [emailprotected]

Direct Purchase Report @ https://www.orbisresearch.com/contact/purchase-single-user/4718175

Follow this link:
Stem Cell Assay Market Report 2020 by Global Key Players, Types, Applications, Countries, Market Size, Forecast to 2026 (Based on 2020 COVID-19...

Protalix BioTherapeutics Appoints Yael Hayon, Ph.D. as its New Vice President, Research and Development – BioSpace

CARMIEL,Israel, June 8, 2020 /PRNewswire/ -- Protalix Biotherapeutics, Inc., (NYSE American: PLX) (TASE: PLX) today announced the appointment of Yael Hayon, Ph.D. as the Company's new Vice President, Research and Development, effective July5, 2020. On June 2, 2020, Yoseph Shaaltiel, Ph.D. retired from his position as the Company's Executive Vice President, Research and Development, effective June 15, 2020.

"Yossi's incredible scientific and entrepreneurial vision led to his founding of Protalix," said Zeev Bronfeld, Chairman of Protalix's Board of Directors. "Yossi's efforts resulted in the development of ProCellEx, our proprietary plant cell-based protein expression system which we use to produce taliglucerase alfa, an approved treatment for Gaucher disease, pegunigalsidase alfa, our investigationaltreatment for Fabry disease which is in the latter stages of clinical development and our other investigationaldrug candidates. The Board of Directors and I are immensely grateful to Yossi for his knowledge, leadership, integrity and professionalism in building Protalix from its founding days to where it is today. We wish him all the best in his future endeavors."

"I am delighted that Yael is joining the Protalix team where she will bring valuable and diverse research& development experience and knowledge," said Dror Bashan, Protalix's President and Chief Executive Officer. "We are greatly thankful to Yossi for his exceptional efforts in founding and building Protalix, and wish him great success in the future."

Dr. Hayon brings to the Company over a decade of experience in pharmaceutical researchand development, both in the scientific operations and the administrative functions. She most recently served as Vice President of Clinical Affairs of Syqe Medical Ltd., Tel-Aviv, where she, among other things, established the clinical and medical global strategy, and was responsible for providing strategic input on the regulatory development plan. Prior to her role at Syqe Medical, Dr. Hayon served as the Head of R&D Israeli Site of LogicBio Therapeutics, Inc., Cambridge, Massachusetts, where she managed LogicBio's Israeli-based Research and Development facility and was involved in strategic decision-making. From 2014 through 2016 she served as the R&D Manager, Stem Cell Medicine Ltd., Jerusalem, Israel. Dr. Hayon holds a Ph.D. in Neurobiology/Hematology, and an MS.c. in Neurobiology, both from the Hebrew University Faculty of Medicine, Jerusalem, Israel.

About Protalix BioTherapeutics, Inc.

Protalix is a biopharmaceutical company focused on the development and commercialization of recombinant therapeutic proteins expressed through its proprietary plant cell-based expression system, ProCellEx. Protalix was the first company to gain U.S.Food and Drug Administration (FDA) approval of a protein produced through plant cell-based in suspension expression system. Protalix's unique expression system represents a new method for developing recombinant proteins in an industrial-scale manner.

Protalix's first product manufactured by ProCellEx, taliglucerase alfa, was approved for marketing by the FDA in May 2012 and, subsequently, by the regulatory authorities of other countries. Protalix has licensed to Pfizer Inc. the worldwide development and commercialization rights for taliglucerase alfa, excluding Brazil, where Protalix retains full rights.

Protalix's development pipeline consists of proprietary versions of recombinant therapeutic proteins that target established pharmaceutical markets, including the following product candidates: pegunigalsidase alfa, a modified version of the recombinant human GalactosidaseA protein for the proposed treatment of Fabry disease; OPRX106, an orally-delivered anti-inflammatory treatment; alidornase alfa for the treatment of Cystic Fibrosis; and others. Protalix has partnered with Chiesi Farmaceutici S.p.A., both in the United States and outside the United States, for the development and commercialization of pegunigalsidase alfa.

Forward-Looking Statements

To the extent that statements in this press release are not strictly historical, all such statements are forward-looking, and are made pursuant to the safe-harbor provisions of the Private Securities Litigation Reform Act of 1995. The terms "expect," "anticipate," "believe," "estimate," "project," "plan," "should" and "intend," and other words or phrases of similar import are intended to identify forward-looking statements. These forward-looking statements are subject to known and unknown risks and uncertainties that may cause actual future experience and results to differ materially from the statements made. These statements are based on our current beliefs and expectations as to such future outcomes. Drug discovery and development involve a high degree of risk and the final results of a clinical trial may be different than the preliminary findings for the clinical trial. Factors that might cause material differences include, among others: that the FDA might not grant marketing approval for PRX102 in the currently anticipated timeline or at all and, if approved, whether PRX102 will be commercially successful; failure or delay in the commencement or completion of our preclinical and clinical trials; risks associated with the novel coronavirus disease (COVID19) outbreak, which may adversely impact our business, preclinical studies and clinical trials; the inherent risks and uncertainties in developing drug platforms and products of the type we are developing; the impact of development of competing therapies and/or technologies by other companies and institutions; and other factors described in our filings with the U.S.Securities and Exchange Commission. The statements in this press release are valid only as of the date hereof and we disclaim any obligation to update this information, except as may be required by law.

Investor ContactChuck Padala, Managing DirectorLifeSci Advisors+1-646-627-8390chuck@lifesciadvisors.com

Media ContactBrian PinkstonLaVoieHealthScience+1-857-588-3347bpinkston@lavoiehealthscience.com

View original content to download multimedia:http://www.prnewswire.com/news-releases/protalix-biotherapeutics-appoints-yael-hayon-phd-as-its-new-vice-president-research-and-development-301072291.html

SOURCE Protalix BioTherapeutics, Inc.

Visit link:
Protalix BioTherapeutics Appoints Yael Hayon, Ph.D. as its New Vice President, Research and Development - BioSpace

COVID-19 Impact and Recovery Analysis – Hematopoietic Stem Cells Transplantation (HSCT) Market 2020-2024 | Demand for Personalized Medicine to Boost…

LONDON--(BUSINESS WIRE)--Technavio has been monitoring the hematopoietic stem cells transplantation (HSCT) market and it is poised to grow by USD 4.64 billion during 2020-2024, progressing at a CAGR of almost 6% during the forecast period. The report offers an up-to-date analysis regarding the current market scenario, latest trends and drivers, and the overall market environment.

Technavio suggests three forecast scenarios (optimistic, probable, and pessimistic) considering the impact of COVID-19. Request for Technavio's latest reports on directly and indirectly impacted markets. Market estimates include pre- and post-COVID-19 impact on the Hematopoietic Stem Cells Transplantation (HSCT) Market. Download free sample report

The market is fragmented, and the degree of fragmentation will accelerate during the forecast period. AllCells Corp., bluebird bio Inc., FUJIFILM Holdings Corp., Lineage Cell Therapeutics Inc., Lonza Group Ltd., MEDIPOST Co. Ltd., Merck & Co. Inc., Sanofi, Takeda Pharmaceutical Co. Ltd., and ThermoGenesis Holdings Inc. are some of the major market participants. To make the most of the opportunities, market vendors should focus more on the growth prospects in the fast-growing segments, while maintaining their positions in the slow-growing segments.

Buy 1 Technavio report and get the second for 50% off. Buy 2 Technavio reports and get the third for free.

View market snapshot before purchasing

Demand for personalized medicine has been instrumental in driving the growth of the market.

Technavio's custom research reports offer detailed insights on the impact of COVID-19 at an industry level, a regional level, and subsequent supply chain operations. This customized report will also help clients keep up with new product launches in direct & indirect COVID-19 related markets, upcoming vaccines and pipeline analysis, and significant developments in vendor operations and government regulations. https://www.technavio.com/report/report/hematopoietic-stem-cells-transplantation-market-industry-analysis

Hematopoietic Stem Cells Transplantation (HSCT) Market 2020-2024: Segmentation

Hematopoietic Stem Cells Transplantation (HSCT) Market is segmented as below:

To learn more about the global trends impacting the future of market research, download a free sample: https://www.technavio.com/talk-to-us?report=IRTNTR41656

Hematopoietic Stem Cells Transplantation (HSCT) Market 2020-2024: Scope

Technavio presents a detailed picture of the market by the way of study, synthesis, and summation of data from multiple sources. The hematopoietic stem cells transplantation (HSCT) market report covers the following areas:

This study identifies the availability of technologically advanced equipment as one of the prime reasons driving the hematopoietic stem cells transplantation (HSCT) market growth during the next few years.

Register for a free trial today and gain instant access to 17,000+ market research reports.

Technavio's SUBSCRIPTION platform

Hematopoietic Stem Cells Transplantation (HSCT) Market 2020-2024: Key Highlights

Table of Contents:

Executive Summary

Market Landscape

Market Sizing

Five Forces Analysis

Market Segmentation by Type

Customer Landscape

Geographic Landscape

Drivers, Challenges, and Trends

Vendor Landscape

Vendor Analysis

Appendix

About Us

Technavio is a leading global technology research and advisory company. Their research and analysis focus on emerging market trends and provides actionable insights to help businesses identify market opportunities and develop effective strategies to optimize their market positions. With over 500 specialized analysts, Technavios report library consists of more than 17,000 reports and counting, covering 800 technologies, spanning across 50 countries. Their client base consists of enterprises of all sizes, including more than 100 Fortune 500 companies. This growing client base relies on Technavios comprehensive coverage, extensive research, and actionable market insights to identify opportunities in existing and potential markets and assess their competitive positions within changing market scenarios.

Read the original:
COVID-19 Impact and Recovery Analysis - Hematopoietic Stem Cells Transplantation (HSCT) Market 2020-2024 | Demand for Personalized Medicine to Boost...

Biobanking Market to reach US $1,801.7 million by 2025 Global Insights on Key Stakeholders, Value Chain Analysis, Growth Drivers, Regulatory…

Dallas, Texas, June 10, 2020 (GLOBE NEWSWIRE) -- The Global Biobanking Market Size 2018, By Specimen Type (Blood Products, Solid Tissue, Cell Lines, Others) Storage Type (Manual Storage, Automated Storage) Application (Regenerative Medicine, Life Science Research, Clinical Research) Region and Forecast 2019 to 2025 study provides an elaborative view of historic, present and forecasted market estimates.

Adroit Market Research report on global biobanking market gives a holistic view of the market from 2015 to 2025, which includes factors such as market drivers, restraints, opportunities and challenges. The market has been studied for historic years from 2015 to 2017, with the base year of estimation as 2018 and forecast from 2019 to 2025. The report covers the current status and future traits of the market at global as well as country level. In addition, the study also assesses the key players based on their product portfolio, geographic footprint, strategic initiatives and overall revenue. Prominent players operating in the global biobanking market have been studied in detail.

Request a pdf sampleathttps://www.adroitmarketresearch.com/contacts/request-sample/1472

The global biobanking market is projected to reach USD 1,801.7 million by 2025, growing at a CAGR of 5.7%. Increase in population genetics studies, advances in biobanking with ongoing research on stem cells and public opting for its preservation, uptake of personalized medicine, government & private funding to provision regenerative medicine research, and the use of genetic information in food safety, forensics, and disease surveillance are the factors driving the growth of the biobanking market.

Biobanks consists of human tissues, DNA, body fluids, for research, therapeutic uses, and biological applications. The demand for biobanks and tissue suppliers have grown exponentially in both numbers and size, and are now established key partners for both academic and commercial groups. A recent study of 456 biobanks in the US showed that nearly two thirds of the biobanks were established within the last decade and 17% have been in existence for over 20 years, with 88% of these part of at least one or more larger organizations (67% academic, 23% hospitals, and 13% research institutes). To sustain this level of growth, biobanks have had to understand and satisfy the different interests of their customers in a sustainable method for long-term success. This move away from repository-like organizations and archive libraries is also evident in the level of donor information that is now collected with samples.

Browse the full report with Tabe of Contents and List of Figures athttps://www.adroitmarketresearch.com/industry-reports/biobanking-market

Biobanks catalogue samples using donor demographics such as age, gender, and ethnicity and may also have information on medical history, genetic traits, environmental factors, and follow-up information. To researchers, this information has become as important as the sample itself and is often a key requirement when sourcing material.

The global biobanking market is categorized based on sample type, storage type and application. Based on storage type the market is segmented into manual storage and automated storage. Manual storage held the largest market share in 2018, while the automated storage segment is likely to grow at the highest CAGR during the forecast period. The benefits of automated storage over manual storage include, reduced labor requirements and costs, improved floor space utilization, increased picking accuracy (reduced picking errors), tighter inventory control, improved picking throughput (speed), and improved ergonomics.

Direct purchase the single user copy athttps://www.adroitmarketresearch.com/researchreport/purchase/1472

In 2018, North America was the largest market for biobanking. Advances in biomedical, pharmaceutical, and biotechnology industries are some of the factors propelling market growth in this region. Key players of the biobanking market include Thermo Fisher Scientific Inc., Tecan Group Ltd., Qiagen N.V., Hamilton Company, Brooks Automation, TTP Labtech Ltd., VWR Corporation, Promega Corporation, Worthington Industries, Chart Industries, Becton, Dickinson and Company, Merck KGaA and Micronic among others.

Are you ooking for a DISCOUNT? then get in touch with us athttps://www.adroitmarketresearch.com/contacts/discount/1472

Major points from Table of Contents:1. Introduction2. Research Methodology3. Market Outlook4. Biobanking Market by Specimen Type, 2015-2025 (USD Million)5. Biobanking Market by Storage Type, 2015-2025 (USD Million)6. Biobanking Market by Application, 2015-2025 (USD Million) 7. Biobanking Market by Region 2015-2025 (USD Million)8. Competitive Landscape9. Company Profiles10. Appendix

Access research repository of Upcoming Reports @ https://adroitmarketresearch.com/upcoming.html

About Us:Adroit Market Research is a global business analytics and consulting company incorporated in 2018. Our target audience is a wide range of corporations, manufacturing companies, product/technology development institutions and industry associations that require understanding of a markets size, key trends, participants and future outlook of an industry. We intend to become our clients knowledge partner and provide them with valuable market insights to help create opportunities that increase their revenues. We follow a code Explore, Learn and Transform. At our core, we are curious people who love to identify and understand industry patterns, create an insightful study around our findings and churn out money-making roadmaps.

Contact Us:Ryan JohnsonAccount Manager - Global3131 McKinney Ave Ste 600Dallas, TX 75204Email ID: sales@adroitmarketresearch.com Phone No.: +1 972-362 -8199Connect with us: Facebook | Twitter | LinkedIn

Go here to see the original:
Biobanking Market to reach US $1,801.7 million by 2025 Global Insights on Key Stakeholders, Value Chain Analysis, Growth Drivers, Regulatory...

Senolytic drugs: can this antibiotic treat symptoms of ageing? – Health Europa

Professor Michael P Lisanti, Chair in Translational Medicine at the University of Salford, has been an active research scientist for more than 30 years and is an expert in the field of cellular senescence. In 2018 Lisanti, along with his wife and research partner Professor Federica Sotgia, co-authored a paper entitled Azithromycin and Roxithromycin define a new family of senolytic drugs that target senescent human fibroblasts, which identified the FDA-approved antibiotic azithromycin as a senolytic drug: a compound which can be used to treat the symptoms of ageing.

Their research was made possible through generous funding contributions from Lunella Biotech, Inc, a Canadian-based pharmaceutical developer which fosters medical innovation; the Foxpoint Foundation, also based in Canada; and the Healthy Life Foundation, a UK charity which funds research into ageing and age-related conditions. Lisanti speaks to HEQ about his work and the future of senescence studies.

We started out focusing on cancer, but the relationship between cancer and ageing led us to shift our focus towards senescence, the process by which cells chronologically age and go into cell cycle arrest. Senescence leads to chronic inflammation: the cells secrete a lot of inflammatory mediators, which allows the cells to become almost infectious; so then neighbouring normal cells become senescent it has a kind of cataclysmic effect. As you age especially as you approach around 50 you begin to accumulate more senescent cells, which are thought to be the root cause of ageing; this then leads to various ageing-associated diseases, such as heart disease, diabetes, dementia and cancer, the most life threatening conditions in the Western world.

The goal, therefore, would be to remove the senescent cells. It is possible to use a genetic trick to remove senescent cells from mice: this causes them to live longer by preventing ageing-associated diseases; but it is not possible to use the same genetic trick for humans. We would therefore need a drug that only kills or removes senescent cells; and that could then potentially lead to rejuvenation, thereby extending the patients healthy lifespan.

We set up a drug assay using normal, commercially available, human fibroblasts: MRC-5, which comes from the lungs, and BJ-1, which comes from the skin. The idea was to artificially induce ageing, which we did using a compound called BrdU. This compound is a nucleoside: it incorporates into the DNA and that leads to DNA damage; and the DNA damage in turn induces the senescence phenotype. The overarching concept was to create a population of cells artificially that were senescent; and then to compare primary cells that were normal with cells which were senescent, with the goal of identifying drugs which could only selectively kill the senescent cells and not harm the normal cells.

We had previously observed positive results in tests on the metabolic effects of antibiotics, so our drug screening identified two drugs called azithromycin and roxithromycin, which constitute a new family of senolytic drugs. Theyre both clinically approved drugs azithromycin has been around longer; and has a strong safety profile and we looked at other members of the same drug family such as erythromycin, which is the parent compound, but erythromycin has no senolytic activity. The characteristics we were looking for appeared to be relatively restricted to azithromycin, which in our observation was very efficiently killing the senescent cells. As we reported in the paper, it had an efficacy of approximately 97%, meaning that it was able to facilitate the growth of the normal cells, while concurrently selectively killing the senescent cells.

We tested the drug on normal and senescent cells which were otherwise identical. The senescent cells underwent apoptosis programmed cell death so that led us to the conclusion that the drug selectively kills the senescent cells, while at the same time the normal cells are able to continue to proliferate. That selective effect of removing exclusively the senescent cells is what we were searching for; because in this instance we would want a drug that could potentially be used in humans and which would only kill senescent cells.

Obviously, we would have to do clinical trials going forward, but the first step should be to identify the pharmaceutical application. Given that this drug appears to selectively kill and remove the senescent cells, it could be used potentially to prevent ageing-associated disease; and it could therefore potentially extend the human lifespan, especially in terms of reducing diseases and conditions like diabetes, heart disease, dementia and even cancer.

Cystic fibrosis is the most common genetic disease in humans; patients with cystic fibrosis are prone to bacterial lung infections. Researchers started to explore the possibility of using azithromycin preventatively in patients with cystic fibrosis; and they found that, while it didnt necessarily affect patients susceptibility to infection, it did prevent lung fibrosis where the lungs become stiff and the patient is unable to breathe and in doing so, extended the patients lifespan. These studies were focused on myofibroblasts, which at the time werent really seen as senescent; whereas the literature now acknowledges a general consensus that myofibroblasts are indeed senescent cells.

We havent specifically examined anything relating ageing to antimicrobial resistance; but azithromycin is an antibiotic, which is not ideal within the context of AMR. Potentially in the future, once researchers identify what it is about the azithromycin that is causing the senescent cells to die, they could develop future drugs azithromycin is a stepping stone in this context, but what it shows is proof of principle that a drug can be identified which selectively kills senescent cells. This indicates that senescent cells are clearly biochemically distinct from the normal cells, and that it is possible to find a drug that selectively kills them and that is relatively safe. It provides a starting point for further new drug discovery to identify other drugs which might also be selective.

Ideally, we would want a drug which is not an antibiotic; but that means further research will be necessary to find additional drugs or to refine the senolytic activity which weve discovered in this drug. We are in the early stages; the point is that it is experimentally feasible and this would then lend itself to doing new clinical trials in the future, because azithromycin is relatively safe and it probably wont need to be administered over a long period of time to remove senescent cells you might not need to use it for any longer than you would as an antibiotic.

This research has been supported by the Foxpoint Foundation (Canada), the Healthy Life Foundation (UK), and Lunella Biotech, Inc. (Canada).

Professor Michael P Lisanti is Chair of Translational Medicine at the University of Salford School of Science, Engineering & Environment, UK. His current research programme is focused on eradicating cancer stem cells (CSCs); and anti-ageing therapies, in the context of age-associated diseases, such as cancer and dementia.

Lisanti began his education at New York University, US, graduating magna cum laude in chemistry (1985); before completing an MD-PhD in cell biology and genetics at Cornell University Medical College, US (1992). In 1992, he moved to MIT, US, where he worked alongside Nobel laureate David Baltimore and renowned cell biologist Harvey Lodish as a Whitehead Institute fellow (1992-96).

His career has since taken him to the Albert Einstein College of Medicine, US (1997-2006), the Kimmel Cancer Center, US (2006-12), and the University of Manchester, UK (2012-16), where he served as the Muriel Edith Rickman chair of breast oncology, director of the Breakthrough Breast Cancer and the Breast Cancer Now Research Units, and founder and director of the Manchester Centre for Cellular Metabolism.

Lisanti has contributed to 564 publications in peer-reviewed journals and been cited more than 90,000 times. A list of his works can be found at: https://pubmed.ncbi.nlm.nih.gov/?term=lisanti+mp&sort=date

Professor Federica Sotgia currently serves as chair in cancer biology and ageing at the University of Salford School of Science, Engineering and Environment, UK, where she focuses on, inter alia, the role of the tumour microenvironment in cancer and the metabolic requirements of tumour-initiating cells.

Sotgia graduated magna cum laude with an MS in biological sciences (1996) from the University of Genova, Italy, where she later completed a PhD in medical genetics (2001). She moved to the Albert Einstein College of Medicine, US, in 1998, originally as a visiting student and then postdoctoral fellow, and she was appointed an instructor in 2002.

Sotgia has since worked as an assistant professor at the Kimmel Cancer Center, US (2006-12), a senior lecturer at the University of Manchester, UK (2012-16), and a Professor in biomedical science at the University of Salford (2016-present).

She has contributed to 206 publications in peer-reviewed journals and been cited upwards of 27,000 times.

A list of her works can be found at: https://pubmed.ncbi.nlm.nih.gov/?term=sotgia+f&sort=date

Professor Michael P Lisanti, MD-PhD, FRSA, FRSBChair in Translational MedicineSchool of Science, Engineering & EnvironmentUniversity of Salford+44 (0)1612 950 240M.P.Lisanti@salford.ac.uk

This article is from issue 13 of Health Europa. Clickhere to get your free subscription today.

More:
Senolytic drugs: can this antibiotic treat symptoms of ageing? - Health Europa

Global Regenerative Medicine Market | Industry Outlines, Growth, Trends, In-Depth Analysis And Outlook Till 2026 – Cole of Duty

The global Regenerative Medicine Market is projected to grow with a striking growth rate of 24.2 % over the forecast period 20192026 divulges the latest research report presented by Big Market Research.

The report represents a basic overview of the market status, competitor segment with a basic introduction of key vendors, top regions, product types and end industries. This report gives a historical overview of the market trends, growth, revenue, capacity, cost structure, and key drivers analysis.

The report is an exhaustive analysis of this market across the world. It offers an overview of the market including its definition, applications, key drivers, key market players, key segments, and manufacturing technology. In addition, the study presents statistical data on the status of the market and hence is a valuable source of guidance for companies and individuals interested in the industry. Additionally, detailed insights on the company profile, product specifications, capacity, production value, and market shares for key vendors are presented in the report.

Request a sample of this premium research: https://www.bigmarketresearch.com/request-sample/3199901?utm_source=SHASHI&utm_medium=COD

The total market is further segmented based on company, country, and application/type for competitive landscape analysis. On the contrary, information on industry chain structure, emerging applications, and technological developments in the market makes the report a must-read document.

The report reveals detailed information about the global key players as well as some small players of the Regenerative Medicine sector.

Target Audience of the Global Regenerative Medicine Market in Market Study:Key Consulting Companies & AdvisorsLarge, medium-sized, and small enterprisesVenture capitalistsValue-Added Resellers (VARs)Third-party knowledge providersInvestment bankersInvestors

These insights help determine the strength of competition and take the necessary steps to obtain a leading position in the Regenerative Medicine industry.

Additionally, the research provides a detailed analysis of the key segments of the market with the help of charts and tables. An overview of each market segment such as type, application, and region are also provided in the report. These insights help in understanding the global trends in the Regenerative Medicine industry and form strategies to be implemented in the future.

The regional analysis of global Regenerative Medicine market is considered for the key regions such as Asia Pacific, North America, Europe, Latin America and Rest of the World. North America is the leading/significant region across the world in terms of market share owing to the high disposable income coupled with rising trend of interior designing in the region. Whereas, Asia-Pacific is also anticipated to exhibit highest growth rate / CAGR over the forecast period 2019-2026

Our analysis involves the study of the market taking into consideration the impact of the COVID-19 pandemic. Please get in touch with us to get your hands on exhaustive coverage of the impact of the current situation on the market. Our expert team of analysts will provide as per report customized to your requirement. For more connect with us at [emailprotected] or call toll free: +1-800-910-6452

Request a discount on standard prices of this premium research: https://www.bigmarketresearch.com/request-for-discount/3199901?utm_source=SHASHI&utm_medium=COD

Key Market Segments

The key players profiled in this report include: AcelityL.P.Inc., NuvasiveInc., VericelCorporation, OsirisTherapeuticsInc., StrykerCorporation, MedtronicPLC

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:

The key product type of Regenerative Medicine market are: Stem Cell Therapy, Biomaterial, Tissue Engineering, Others

The study clearly reveals that the Regenerative Medicine industry has attained remarkable growth since 2019-2026. This research report is prepared based on an in-depth analysis of the market by experts. As a final point, stakeholders, investors, product managers, marketing executives, and other professionals seeking unbiased data on supply, demand, and future forecasts would find the report valuable.

Table of Contents

Chapter 1. Global Regenerative Medicine Market Definition and ScopeChapter 2. Research MethodologyChapter 3. Executive SummaryChapter 4. Global Regenerative Medicine Market DynamicsChapter 5. Regenerative Medicine Market, by ComponentChapter 6. Global Regenerative Medicine Market, by ServicesChapter 7. Global Regenerative Medicine Market, by Organization SizeChapter 8. Regenerative Medicine Market, by VerticalChapter 9. Regenerative Medicine Market, by Regional AnalysisChapter 10. Competitive Intelligence

About Us:

Big Market Research has a range of research reports from various publishers across the world. Our database of reports of various market categories and sub-categories would help to find the exact report you may be looking for.

We are instrumental in providing quantitative and qualitative insights on your area of interest by bringing reports from various publishers at one place to save your time and money. A lot of organizations across the world are gaining profits and great benefits from information gained through reports sourced by us.

Contact us:Mr. Abhishek Paliwal5933 NE Win Sivers Drive, #205, Portland,OR 97220 United StatesDirect: +1-971-202-1575Toll Free: +1-800-910-6452Email [emailprotected]

Excerpt from:
Global Regenerative Medicine Market | Industry Outlines, Growth, Trends, In-Depth Analysis And Outlook Till 2026 - Cole of Duty