Global Animal Stem Cell Therapy Market To Reach A New Threshold of Growth By 2026 – The Courier

There are millions of them around the globe waiting for clutching on to some of the latest vital information circulating across the globe. The up-to-the-minute Animal Stem Cell Therapy market report based on the growth and the development of theAnimal Stem Cell Therapy marketis systematically listed down. The Animal Stem Cell Therapy market report comprises statistically verified facts such the unique essence including topological investigations, worldwide market share, government stringent norms, applications, current trends, futuristic plans, market bifurcations, and so on mentioned in a crystal clear pattern.

The statistical plus scientific Animal Stem Cell Therapy market report has all the important market aspects penciled down in a layman language format so that the data based on the markets productivity or future strategy can be easily extrapolated from the reports. The Animal Stem Cell Therapy market report has the dominant market players ANIMAL CELL THERAPIES, Celavet, Animacel, VETSTEM BIOPHARMA, Cell Therapy Sciences, Magellan Stem Cells, Cells Power Japan, Animal Care Stem, Aratana Therapeutics, VetCell Therapeutics, MediVet Biologic, U.S. Stem Cell, J-ARM explained in detail.

Sample of global Animal Stem Cell Therapy Market Report at::https://www.marketresearchstore.com/report/global-animal-stem-cell-therapy-market-report-2020-751651#RequestSample

The essential futuristic segments such as {Dogs, Horses, Others}; {Veterinary Hospitals, Research Organizations} have also been detailed out in the Animal Stem Cell Therapy market report for the clients convenience and more of vital data embracing capability. The forecast trends along with the current market status can better understand the Animal Stem Cell Therapy market development on a global basis. The intricate industrial strategies and the supply-demand chain are also discussed in the contextual report.

Key points of the global Animal Stem Cell Therapy market

Theoretical analysis of the global Animal Stem Cell Therapy market stimulators, products, and other vital facets Recent, historical, and future trends in terms of revenue and market dynamics are reported Pin-point analysis of the competitive market dynamics and investment structure is predicted to grow Future market trends, latest innovations, and various business strategies are reported Market dynamics include growth influencers, opportunities, threats, challenges, and other crucial facets

The Animal Stem Cell Therapy market report has the imperative data mentioned in a systematic way only after comprehensive inspection and vigilant referencing. The diagrammatic representations such as a pie chart of the Animal Stem Cell Therapy market are also drawn out so as to attract the punters and make it easy for them to comprehend the entire Animal Stem Cell Therapy market from in and out. The most eye-catching format of the Animal Stem Cell Therapy market report is its market bifurcation based on the product type, application, geography, end-users, and more as per the particular market. The geographical segments Europe, North America, Latin America, Asia Pacific, and Middle East & Africa are further exhaustively mentioned.

Read Detailed Index of full Research Study at::https://www.marketresearchstore.com/report/global-animal-stem-cell-therapy-market-report-2020-751651

Questions answered in the report include

1. What is the expected market size by the end of the forecast period? 2. What are the major factors initiating the global Animal Stem Cell Therapy market growth? 3. What are the latest developments and trending market strategies that are influencing the growth of the Animal Stem Cell Therapy market? 4. What are the key outcomes of the Animal Stem Cell Therapy market developments? 5. Who are the key players in the market? 6. What are the opportunities and challenges faced by the key players?

The Animal Stem Cell Therapy market report endows the global market dominance, market segmentation, growth factors, and others reported such that the clients can have a total tour of the market without any much off efforts needed.

Key Reasons for Purchasing Global Animal Stem Cell Therapy Market Report

New approaches and latest development trend that describe the structure of the market Advanced market breakdown structure Historical data and future market scope In-depth market analysis based on statistics, growth stimulators, and market developments Statistical data representation through figurative, numerical, and theoretical elaboration Report provides insight of the business and sales activities

Ask Any Kind of Query Before Purchasing the Report Here:https://www.marketresearchstore.com/report/global-animal-stem-cell-therapy-market-report-2020-751651#InquiryForBuying

Visit link:
Global Animal Stem Cell Therapy Market To Reach A New Threshold of Growth By 2026 - The Courier

Cryo-shocked cancer cells for targeted drug delivery and vaccination – Science Advances

Abstract

Live cells have been vastly engineered into drug delivery vehicles to leverage their targeting capability and cargo release behavior. Here, we describe a simple method to obtain therapeutics-containing dead cells by shocking live cancer cells in liquid nitrogen to eliminate pathogenicity while preserving their major structure and chemotaxis toward the lesion site. In an acute myeloid leukemia (AML) mouse model, we demonstrated that the liquid nitrogentreated AML cells (LNT cells) can augment targeted delivery of doxorubicin (DOX) toward the bone marrow. Moreover, LNT cells serve as a cancer vaccine and promote antitumor immune responses that prolong the survival of tumor-bearing mice. Preimmunization with LNT cells along with an adjuvant also protected healthy mice from AML cell challenge.

Acute myeloid leukemia (AML) is a hematological malignancy with a dismal prognosis and 5-year survival of only 30% (1, 2). The standard-of-care cytoreductive chemotherapy induces AML remission (35), but disease relapse frequently occurs (6, 7). Hematopoietic stem cell transplantation (HSCT) in patients who achieve remission after chemotherapy represents the only curative approach so far (8, 9). However, HSCT is associated with either the lack of suitable hematopoietic stem cell donors or the high risk of transplantation-related mortality (10). Hence, there is an urgent need to find further strategies for AML treatment.

AML originates in the bone marrow, and bone marrow creates leukemia-niches that promote leukemia survival (11). Furthermore, biodistribution of chemotherapeutics to the bone marrow is frequently poor (12, 13), and higher doses of chemotherapy required to ablate leukemia are toxic to normal tissues. Thus, developing targeting drug delivery to the bone marrow may not only enhance the therapeutic index of chemotherapy but also reduce its toxicity to nonhematopoietic tissues. Nevertheless, it is still challenging to engineer bone marrowtargeting moieties and bypass the bloodbone marrow barriers (14). Leveraging cells intrinsic properties offers solutions to overcome these limitations (1517). Because AML cells naturally exhibit bone marrow homing capabilities (1820), we developed an approach to use AML cells as drug carriers while eliminating their intrinsic pathogenicity.

Here, we used a liquid nitrogenbased cryo-shocking method to obtain therapeutic dead cells. These cells maintained the intact structure allowing for drug encapsulation, but lost their proliferation ability and pathogenicity. Specifically, cryo-shocked AML cells kept their bone marrow homing capability and served as a drug delivery vehicle of doxorubicin (DOX), which is a critical drug used in the induction chemotherapy in AML. Cryo-shocked AML cells stimulated an immune response that was in conjunction with chemotherapy to eradicate leukemia in tumor-bearing mice. Preimmunization with LNT cells together with an adjuvant protected healthy mice from AML cell challenge. We thus proposed a dead cellbased delivery vehicle that can be rapidly manufactured for clinical use compared with other live cellbased drug delivery systems (21).

To obtain the liquid nitrogentreated (LNT) cells, AML cells were suspended in the cell cryopreservation medium and immersed in liquid nitrogen for 12 hours. LNT cells were then thawed at 37C and washed with phosphate-buffered saline (PBS) (Fig. 1A). When analyzed by confocal microscopy, LNT cells showed the same cellular structure as untreated live cells when assessed by nucleus and cytoskeleton staining (Fig. 1B). A slight decrease in cellular size was observed (Fig. 1C), with an average size of 11 m for LNT cells and 12 m for untreated live cells. The forward scatter (FSC) values measured by flow cytometry corroborated the cell size reduction of LNT cells, and similar side scatter (SSC) values suggested that the internal structure of LNT cells was maintained (Fig. 1D). Scanning electron microscopy (SEM) images revealed the sphere-like structure of LNT cells and the rougher cellular surface as compared with control live cells (Fig. 1E and fig. S1).

(A) Schematic of the procedure to prepare LNT cells. (B) Cellular structure of live and LNT C1498 cells. Cell nucleus was stained by Hoechst 33342 (blue), and cytoplasm F-actin was stained by AF488 phalloidin (green). Scale bars, 10 m. (C) Cellular sizes of live and LNT C1498 cells. The cells were imaged by confocal microscopy, and cellular size was measured by the software Nano Measurer (cell numbers = 200). (D) Flow cytometry analysis of live and LNT C1498 cells under same voltages. FSC, forward scatter; SSC, side scatter. (E) SEM images of live and LNT cells. Scale bars, 1 m. (F) Cell viability analysis of live and LNT cells by LIVE/DEAD viability kit. Calcein AM: live cells; EthD-1: dead cells. Scale bar, 10 m. (G) Cell viability analysis of live and LNT cells by CCK8 assay (n = 6). a.u., arbitrary unit. (H) In vivo proliferation of 2 106 luciferase tagged live and LNT C1498 cells indicated by the bioluminescence signal (n = 5). (I) Survival of mice after challenge with 2 106 live and LNT tumor cells (n = 5). Typical flow cytometry images (J) and DsRed intensities (K) of peripheral blood 20 days after challenge with live and LNT DsRed tagged C1498 cells (n = 3). MFI, mean fluorescence intensity. Data are presented as means SD (G and K). Statistical significance was calculated via the log-rank (Mantel-Cox) test (I) and ordinary one-way analysis of variance (ANOVA) (K). *P < 0.05, **P < 0.01. NS, not significant.

Next, we evaluated the cell viability of LNT cells. As shown in Fig. 1F, nearly all the LNT cells were labeled with EthD-1 (indicating dead cells) and did not show intact fluorescence signal of calcein AM (indicating live cells). In addition, LNT cells did not show proliferative activity as compared with live cancer cells as measured with cell counting kit-8 (CCK8) assay (Fig. 1G). Furthermore, we confirmed the necrosis-dependent cell death of LNT cells by annexin-Vpropidium iodide (PI) staining (fig. S2). We further verified the absence of pathogenicity of LNT cells in vivo. As shown in Fig. 1H, live C1498 AML cells quickly proliferated in mice and caused 100% death in 31 days, while mice receiving C1498 LNT cells exhibited no detectable bioluminescence signal, and all mice survived for at least 180 days (Fig. 1, H and I). Moreover, we quantitatively analyzed cancer cells in the peripheral blood at day 20 after injection. A notably higher DsRed signal was observed in mice injected with live C1498 cells, indicating a high portion of leukemia cells circulating in the blood, while the DsRed intensity for the mice challenged with LNT cells was similar to that of healthy mice (Fig. 1, J and K).

Leukemia cells exhibit bone marrow homing and resident capabilities, which are at least in part associated with the expression of CXCR4 and CD44 chemokine, two typical adhesion receptors that interact with bone marrow (18, 22, 23). SDSpolyacrylamide gel electrophoresis (PAGE) showed that most of the proteins expressed by live C1498 cells were retained in LNT cells (fig. S3A). CXCR4 and CD44 were detected in both live and LNT cells as assessed by confocal imaging and flow cytometry (Fig. 2, A and B, and fig. S3, B and C). Despite some reduction in expression levels, Western blotting analysis indicated that CXCR4 and CD44 expression were 39 and 60%, respectively, in LNT cells compared with live cells (fig. S3, D and E). The bone marrow homing capacity of LNT cells was also evaluated. Upon intravenous infusion, LNT cells exhibited similar accumulation efficiency in bone barrow compared with live C1498 cells (Fig. 2, C and D, and fig. S4A). Cell signal was notably higher compared with paraformaldehyde-fixed cells, which reflects the loss of bioactivities upon paraformaldehyde fixation (Fig. 2, C and D). LNT cells also distributed in the liver, kidney, and spleen (fig. S4B), and were cleared from the bloodstream within 24 hours (fig. S5).

CXCR4 (A) and CD44 (B) expression of live and LNT C1498 cells analyzed by confocal microscopy (top) and flow cytometry (bottom). Scale bars, 10 m. (C) Fluorescence images of bone isolated 6 hours after injection of cy5.5-labeled live C1498 cells, LNT C1498 cells, and paraformaldehyde-fixed C1498 cells. (D) Fluorescence intensities of the bone of indicated groups (n = 6). (E) Typical confocal image of DOX-loaded LNT cells. Scale bar, 10 m. (F) Cumulative release profile of DOX from LNT cell/DOX (n = 3). (G) Plasma DOX concentration after intravenous injection of free DOX and LNT cell/DOX with DOX dose of 2.5 mg/kg (n = 4). (H) Bone marrow DOX content 3 hours after administration of the drug (n = 3). Data are presented as means SD (D and F to H). Statistical significance was calculated via ordinary one-way ANOVA (D) and Students t test (G and H). *P < 0.05, **P < 0.01, ***P < 0.001.

Because nuclear and cytoplasmic cellular structures are preserved in LNT cells (Fig. 1B), we assessed if these cells can be payload with DOX, via DNA intercalation and the electrostatic interactions between DOX and cytoplasm proteins (2426), and deliver DOX to bone marrow. Briefly, DOX could be loaded into LNT cells via mixing and incubation with a loading capacity of 65 16 g per 1 107 LNT cells (Fig. 2E and fig. S6A). DOX was released from the drug-loaded LNT cells (LNT cell/DOX) in a sustained manner, and 81% of DOX was released within 10 hours (Fig. 2F). We then studied the in vitro cytotoxicity against C1498 cells of free DOX and LNT cell/DOX. The IC50 (median inhibitory concentration) values were 0.32 and 1.05 g/ml, respectively (fig. S6B). Even though free DOX exhibited higher cytotoxicity against C1498 cells in vitro, LNT cell/DOX allowed longer detection of DOX in the blood and higher DOX accumulation within the bone marrow (Fig. 2, G and H). We used murine AML models to evaluate the therapeutic efficacy of LNT cell/DOX. In tumor-bearing C57BL/6J mice, tumor growth was monitored by bioluminescence signals upon treatment (fig. S7, A to C). In this leukemia model, although LNT cells alone exhibited no antitumor effects, LNT cell/DOX treatment reduced the tumor growth compared with control treatments (fig. S7, D to H).

Tumor cell lysates can function as cancer vaccines and initiate tumor-specific immune responses (27, 28). We hypothesized that LNT cells can enhance the antigen uptake and maturation of antigen-presenting cells (APCs). LNT cells cocultured with dendritic cells (DCs) caused their maturation as assessed by up-regulation of CD40, CD80, CD86, and major histocompatibility complex II (MHC-II) (fig. S8A). Moreover, CD4+ T cells and CD8+ T cells increased in the peripheral blood of the mice receiving LNT cells and the adjuvant of monophosphoryl lipid A (MPLA) (fig. S8B). DC maturation and T cell activationrelated cytokines, including interferon- (IFN-), tumor necrosis factor (TNF-), and interleukin-6 (IL-6), were also detected in mice treated with LNT cell and adjuvant (fig. S8C). We next evaluated the antitumor efficacy of LNT cell/DOX with adjuvant in leukemia-bearing mice. As demonstrated in Fig. 3 (A and B), bioluminescence of AML cancer cells increased rapidly in untreated mice, while AML had been partially inhibited after DOX or LNT cell and adjuvant treatment. AML cells were almost completely eliminated in mice treated with LNT cell/DOX and adjuvant up to 21 days after tumor inoculation (Fig. 3B). Quantitative analysis of tumor bioluminescence and survival analysis also demonstrated superior therapeutic activity of LNT cell/DOX combined with adjuvant (Fig. 3, C to E). Increased serum levels of IFN- and TNF- (Fig. 3, F and G), as well as elevation of CD3+ T cell and CD8+ T cells, supported the occurrence of boosted immunity in the mice receiving LNT Cell/DOX and adjuvant treatment (Fig. 3, H and I).

(A) Schematic of the treatment model. (B) AML progression in vivo as indicated by bioluminescence signal expressed by luciferase tagged C1498 cells during different treatments (G1, saline; G2, DOX; G3, LNT cell + adjuvant; G4, LNT cell/DOX + adjuvant). (C) Quantified bioluminescence of different treatment groups. (D) Bioluminescence intensity of treated mice on day 21 (n = 6). (E) Survival of the mice of different treatment groups (n = 6). Serum cytokine levels of IFN- (F), TNF- (G), and proportion of peripheral CD3+ T cells (H) and CD8+ T cells (I) on day 13 (n = 6). Data are presented as means SD. (D and F to I). Statistical significance was calculated via ordinary one-way ANOVA (D and F to I) and log-rank (Mantel-Cox) test (E). *P < 0.05, **P < 0.01, ***P < 0.001.

We further evaluated the efficacy of LNT cells as a prophylactic cancer vaccine. Mice were first immunized at 21, 14, and 7 days before challenge with live C1498 cells. The onset of AML in mice was prevented in mice preimmunized with LNT cells and adjuvant (Fig. 4, A to C). Quantitative data also revealed that the tumor bioluminescence intensity of the group of LNT cells with adjuvant was substantially lower than control groups (Fig. 4D). Moreover, 71% of the mice treated with LNT cells and adjuvant were tumor free 90 days after tumor challenge, while all control mice died by day 34 (Fig. 4E). Serum levels of IFN-, TNF-, IL-12, and IL-6 were significantly increased in mice treated with LNT cells and adjuvant (Fig. 4F), indicating that a prompt immune response was triggered upon tumor cell inoculation. In addition, CD3+ T cells and CD8+ T cells were significantly increased in the peripheral blood of mice vaccinated with LNT cells and adjuvant (Fig. 4, G and H, and fig. S9).

(A) Schematic of the treatment model. Bioluminescence images (B) and quantified bioluminescence (C) of the mice preimmunized with different treatment formulations (G1, saline; G2, adjuvant; G3, LNT cell + adjuvant). (D) Bioluminescence intensity of treated mice on day 47 (n = 5 for G1 and G2 for one mice died before day 47; n = 7 for G3). (E) Survival of the mice after tumor challenge (n = 6 for G1 and G2; n = 7 for G3). (F) Serum cytokine levels 3 days after challenge of live C1498 cells (n = 6 for G1 and G2; n = 7 for G3). (G) Representative flow cytometry images of CD3+ T cells (left) and proportion of peripheral CD3+ T cells (right) on day 24 (n = 6 for G1 and G2; n = 7 for G3). (H) Representative flow cytometry images of CD8+ T cells (left) and corresponding proportion of peripheral CD8+ T cell gating on CD3+ T cells (right) on day 24 (n = 6 for G1 and G2; n = 7 for G3). Data are presented as means SD. (D and F to H). Statistical significance was calculated via ordinary one-way ANOVA (D and F to H) and log-rank (Mantel-Cox) test (E), *P < 0.05, **P < 0.01, ***P < 0.001.

In this study, we demonstrated the feasibility, efficacy, and safety of tumor dead cells used as a drug-targeting carrier and tumor vaccine for cancer therapy. Compared with the synthetic materialmediated delivery vehicles, cell-based carriers show unique targeting capacities and can bypass biological barriers (15, 29). AML cells originate in the bone marrow and naturally exhibit similar bone marrow homing capabilities as HSCs (22, 30, 31), rendering them suitable to be used as cellular drug carriers for AML therapy. However, it remains essential to develop strategies allowing the elimination of AML tumorigenicity while transiently preserving cellular integrity to deliver the payload at the tumor site. We therefore proposed to use the dead but functional AML cells as the drug carrier.

Usually, the structure of the live cells can disintegrate upon dying with the loss of proteins and cytokines (32). In addition, external stimuli that could induce cell death, such as heat or radiation, will deactivate proteins as well (33, 34). Our data support the concept that cryo-shocked tumor cells obtained by rapid immersion of live cells in liquid nitrogen lose tumorigenicity while preserving transiently the integrity of the cell structure, which is critical for the drug loading and cargo release. Furthermore, certain critical functional proteins that include CD44 and CXCR4 were retained in LNT cells. CD44 can interact with hyaluronic acid that is highly expressed in the endosteum of bone marrow (18). CXCR4 enables cells to migrate toward the chemokine stromal cellderived factor 1 (SDF-1) that is constitutively produced by the osteoblasts and stromal cells (23). CD44 and CXCR4 are two important adhesion receptors mediating AML cells homing toward bone marrow (35). Retention of both CD44 and CXCR4 in LNT cells, even if at reduced levels compared with live cells, is likely critical to promote their bone marrow homing. The proposed LNT-based strategy is simple and straightforward from a manufacturing point of view. Tumor cells in the case of liquid tumors can be readily collected in large quantity, for example, by leukapheresis. Similarly, for solid tumors, multiple devises are currently available to generate single cell suspension from resected tumors or tumor biopsies. The process of cell shocking in liquid nitrogen is also feasible to standardize in good manufacturing practice conditions.

We evaluated the proliferation and tumorigenicity of LNT tumor cells both in vitro and in vivo. The data that all mice treated with LNT cells exhibited no obvious side effects and no leukemia growth was recorded for 6 months after inoculation of LNT C1498 cells support at least in our mouse model the safety of the proposed strategy. After exposure to liquid nitrogen, the cellular membrane of LNT cells becomes permeable. While live cells require treatment with cell membrane detergent to obtain intracellular staining, LNT cells do not require this treatment, indicating the loss of long-term integrity of the cell membrane, which is essential to cell survival. However, our experiments demonstrate that liquid nitrogen treatment does not impair the capacity of LNT cells to function as drug carrier and tumor vaccine. The cryo-shocking technique could be a platform technology in cell bioengineering and could be applicable to various cell types. Here, we have further tested feasibility in 4T1 tumor cells (fig. S10). Regarding the potential impact in clinical use, the safety of LNT tumor cells, besides C1498 cells adopted in this work, should be evaluated thoroughly in other experimental animal models. In addition, the application of LNT cells to serve as drug carriers of other therapeutics, such as immune checkpoint inhibitors (36), is worth investigating.

In summary, we engineered LNT tumor cells to serve simultaneously as a drug delivery carrier and cancer vaccine. The simple liquid nitrogen treating process abrogates the tumorigenicity of tumor cells but preserves the integrity of their cellular structure. This in turn allows the possibility to load LNT cells with chemotherapy drugs and preserves the homing capacity of these cells to the tumor site. LNT cells in combination with adjuvant could elicit both therapeutic and protective immune antitumor responses and may avoid the complex quality control associated with isolated cells and synthesized materialbased vehicles and enable large-scale production for clinical use.

The aim of this study was to use the cryo-shocked tumor cells as a kind of drug-targeting carrier and tumor vaccine for chemo-immunotherapy in the treatment of AML. After treating the live cells in liquid nitrogen, the cellular structure of the cryo-shocked cells was observed. The proliferation behavior, in vivo tumorigenicity, and targeting capability toward the bone marrow of the cryo-shocked cells were assessed. In vivo antitumor efficacy was analyzed in an AML model by intravenously injecting C1498 cells in C57BL/6J mice. Mice were randomly assigned to groups based on body weights. After different treatments, the mice were captured by in vivo imaging system (IVIS) to evaluate in vivo tumor progression. Survival curves, immune cell proportions, and cytokine levels were determined according to previous experimental experience. Specific information about treatment groups, sample numbers, and data analysis was denoted in the figure captions.

Doxorubicin hydrochloride was purchased from Fisher Scientific Co. (D4193; purity, >95%). Noncontrolled-rate cell cryopreservation medium was bought from Cyagen Co. (NCRC-10001-50). AML cell line C1498 was purchased from the American Type Culture Collection (ATCC). Luciferase and DsRed tagged C1498 cell line was provided by B. Blazar of the University of Minnesota. The cells were cultured in 90% Dulbeccos modified Eagles medium (Gibco) and 10% fetal bovine serum (Gibco) with penicillin (200 U ml1) and streptomycin (200 U ml1) (Gibco). The cells were passaged every 1 to 2 days. C57BL/6J mice (4 to 6 weeks, female) were purchased from the Jackson laboratory. All animal tests complied with the animal protocol approved by the Institutional Animal Care and Use Committee of the University of California, Los Angeles.

C1498 cells were centrifuged at 250g for 3 min and suspended in noncontrolled-rate cell cryopreservation medium at a cell density of 1 106 to 1 107 ml1. The cell-containing medium was immersed in liquid nitrogen for 12 hours. Before use, the medium was thawed at 37C and LNT cells were pelleted at 500g for 3 min. After washing with PBS solution (pH 7.4), LNT cells were suspended in PBS and kept at 4C. For preparation of DOX-loaded LNT cells, the LNT cells were suspended in DOX containing PBS. After incubation for 2 hours, the medium was centrifuged at 500g for 5 min and the pellets were DOX-loaded LNT cells.

The AML model was established by intravenous injection of 5 106 C1498 cells on day 0. On day 8 and day 15, saline, LNT cell + adjuvant, free DOX, and LNT cell/DOX + adjuvant were administrated intravenously with DOX dose of 5 mg/kg and adjuvant (MPLA) 20 g per mouse. Specifically, MPLA was intravenously injected 10 hours after injection of LNT cell or LNT cell/DOX. The bioluminescence images of mice were captured every 3 days. The exposure time was 2 min. On day 13, 400 l of blood was collected via the orbital vein. Blood (200 l) was treated with ammonium-chloride-potassium (ACK) buffer and centrifuged at 800g for 8 min to obtain pellets of white blood cells. After staining with BV421-CD3, PE-CD4, and APC-CD8, the samples were analyzed by flow cytometry. Another 200 l of blood in blood serum collection tubes (BD Microtainer 365967) was centrifuged at 3000 rpm for 10 min. The upper serum was detected with the following enzyme-linked immunosorbent assay kits: IFN- (BioLegend 430804) and TNF- (BioLegend 430904).

The results were presented as means SD or mean standard error of the mean (means SEM) as indicated. The data were compared by Students t test between two groups and ordinary one-way analysis of variance (ANOVA) for three or more groups. The survival curves were analyzed via the log-rank (Mantel-Cox) test. All statistical analyses were conducted by the GraphPad Prism software. The threshold of a statistically significant difference was defined as P < 0.05.

Acknowledgments: We acknowledge B. Blazar at the University of Minnesota for providing the luciferase and DsRed tagged C1498 cell line. Funding: This work was supported by the NIH (R01 CA234343-01A1) and grants from the start-up packages of UCLA. Author contributions: Z.G. and T.C. proposed the conception of the project. T.C. and H.L. performed all the experiments and collected the data with the help of G.C., Z.W., J.W., P.A., and Y.T. All authors analyzed the data and contributed to the writing of the manuscript, discussed the results and implications, and edited the manuscript at all stages. Competing interests: Z.G. and T.C. are inventors on a U.S. patent application related to this work filed by University of California, Los Angeles (no. 63/094,034, filed [Oct 20th, 2020]). Z.G. is a scientific cofounder of ZenCapsule Inc. The authors declare that they have no other competing interests. Data and materials 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.

See the rest here:
Cryo-shocked cancer cells for targeted drug delivery and vaccination - Science Advances

Global Stem Cells Cryopreservation Equipment Market 2020 forecast to 2028, by types, by applications, by analysis with Worthington Industries, Cesca…

Global Stem Cells Cryopreservation Equipment Market is projected to grow at a CAGR +22% during the forecast period from 2020 to 2028.

Stem cells, which hold the promise of new cell-based therapies to treat critical medical conditions, have to be effectively stored until its time to use them. The method is called cryopreservation: it freezes stem cells at temperatures below -150oC, by immersing them in nitrogen vapour.

Stem cells will one day be effective in the treatment of many medical conditions and diseases. But unproven stem cell treatments can be unsafe so get all of the facts if youre considering any treatment. Stem cells have been called everything from cure-alls to miracle treatments.

Types of Stem Cells:-

Stem cells are divided into 2 main forms. They are embryonic stem cells and adult stem cells. Embryonic stem cells. The embryonic stem cells used in research today come from unused embryos.

Request a Sample Copy of this report:-

https://www.reportconsultant.com/request_sample.php?id=78516

Key Players:-

Global Stem Cells Cryopreservation Equipment Market by Product:-

Global Stem Cells Cryopreservation Equipment Market by Applications:-

Due to the pandemic, we have included a special section on the Impact of COVID 19 on the Stem Cells Cryopreservation Equipment Market which would mention How the Covid-19 is affecting the Stem Cells Cryopreservation Equipment Industry, Market Trends and Potential Opportunities in the COVID-19 Landscape, Covid-19 Impact on Key Regions and Proposal for Stem Cells Cryopreservation Equipment Players to Combat Covid-19 Impact.

A new statistical report has recently published by Reports Consultant to its massive repository titled as Global Stem Cells Cryopreservation Equipment market 2020. This informative document takes a closer and analytical look on different aspects of the businesses to understand the business structure clearly. It has been compiled by using primary and secondary research techniques. Furthermore, it makes use of graphical presentation techniques such as ample graphs, charts, tables, and pictures.

Get up to 40% Discount:-

https://www.reportconsultant.com/ask_for_discount.php?id=78516

Global Stem Cells Cryopreservation Equipment Market by Region analysis:-

Why This Report important?

Global Stem Cells Cryopreservation Equipment Market Table of Content (TOC):-

Chapter1 Introduction

Chapter2 Research Scope

Chapter3 Global Stem Cells Cryopreservation Equipment Market Segmentation

Chapter4 Research Methodology

Chapter5 Definitions and Assumptions

Chapter6 Executive Summary

Chapter7 Global Stem Cells Cryopreservation Equipment Market Dynamics

Chapter8 Global Stem Cells Cryopreservation Equipment Market Key Players

Chapter9 Conclusion

Chapter10 Appendix

About Us:

Report Consultant A worldwide pacesetter in analytics, research and advisory that can assist you to renovate your business and modify your approach. With us, you will learn to take decisions intrepidly by taking calculative risks leading to lucrative businesses in the ever-changing market. We make sense of drawbacks, opportunities, circumstances, estimations and information using our experienced skills and verified methodologies.

Our research reports will give you the most realistic and incomparable experience of revolutionary market solutions. We have effectively steered businesses all over the world through our market research reports with our predictive nature and are exceptionally positioned to lead digital transformations. Thus, we craft greater value for clients by presenting progressive opportunities in the global futuristic market.

Contact us:

(Report Consultant)

Contact No: +81-368444299

sales@reportconsultant.com

http://www.reportconsultant.com

Original post:
Global Stem Cells Cryopreservation Equipment Market 2020 forecast to 2028, by types, by applications, by analysis with Worthington Industries, Cesca...

A Potential Therapy for One of the Leading Causes of Heart Disease – PRNewswire

After 15 years of unrelenting work, a team of scientists from Gladstone Institutes has now discovered a potential drug candidate for heart valve disease that works in both human cells and animals and is ready to move toward a clinical trial. Their findings were just published in the journal Science.

"The disease is often diagnosed at an early stage and calcification of the heart valves worsens over the patient's lifetime as they age," says Gladstone President and Director of the Roddenberry Stem Cell Center Deepak Srivastava, MD,who led the study. "If we could intervene early in life with an effective drug, we could potentially prevent the disease from occurring. By simply slowing the progression and shifting the age of people who require interventions by 5 or 10 years, we could avoid tens of thousands of surgical valve replacements every year."

This also applies to the millions of Americansabout one to two percent of the populationwith a congenital anomaly called bicuspid aortic valve, in which the aortic valve only has two leaflets instead of the normal three. While some people may not even know they have this common heart anomaly, many will be diagnosed as early as their forties.

"We can detect this valve anomaly through an ultrasound," explains Srivastava, who is also a pediatric cardiologist and a professor in the Department of Pediatrics at UC San Francisco (UCSF). "About a third of patients with bicuspid aortic valve, which is a very large number, will develop enough calcification to require an intervention."

Srivastava's research into heart valve disease started in 2005, when he treated a family in Texas who had this type of early-onset calcification. All these years later, thanks to the family's donated cells, his team has finally found a solution to help them and so many others.

A Holistic Approach in the Hunt for a Therapy

Members of the family treated by Srivastava had disease that crossed five generations, enabling the team to identify the causea mutation in one copy of the gene NOTCH1. Mutations in this gene cause calcific aortic valve disease in approximately four percent of patients and can also cause thickening of valves that trigger problems in newborns. In the other 96 percent of cases, the disease occurs sporadically.

"The NOTCH1 mutation provided a foothold for us to figure out what goes wrong in this common disease, but most people won't have that mutation," says Srivastava. "However, we found that the process that leads to the calcification of the valve is mostly the same whether individuals have the mutation or not. The valve cells get confused and start thinking they're bone cells, so they start laying down calcium and that leads to hardening and narrowing of the valves."

In the hunt for a treatment, the group of scientists chose a novel, holistic approach rather than simply focusing on a single target, such as the NOTCH1 gene.

"Our goal was to develop a new framework to discover therapeutics for human disease," says Christina V. Theodoris, MD, PhD, lead author of the study who is now completing her residency in pediatric genetics at Boston Children's Hospital. "We wanted to find promising therapies that could treat the disease at its core, as opposed to just treating some specific symptoms or peripheral aspects of the disease."

When Theodoris first joined Srivastava's lab at Gladstone, she was a graduate student at UCSF. At the time, they knew the NOTCH1 gene mutation caused valve disease, but they didn't have the tools to study the problem further, largely because it was very difficult to obtain valve cells from patients.

"My first project was to convert the cells from the patient families into induced pluripotent stem (iPS) cells, which have the potential of becoming any cell in the body, and turn them into cells that line the valve, allowing us to understand why the disease occurs," says Theodoris. "My second project was to make a mouse model of calcific aortic valve disease. Only then could we start using these models to identify a therapy."

One Drug Candidate Rises to the Top

For this latest study, the scientists searched for drug-like molecules that could correct the overall network that goes awry in heart valve disease and leads to calcification. To do so, they first had to determine the network of genes that are turned on or off in diseased cells.

Then, they used an artificial intelligence method, training a machine learning program to detect whether a cell was healthy or sick based on this network of genes. They subsequently treated diseased human cells with nearly 1,600 molecules to see if any drugs shifted the network in the cells enough that the machine learning program would reclassify them as healthy. The researchers identified a few molecules that could correct diseased cells back to the normal state.

"Our first screen was done with cells that have the NOTCH1 mutation, but we didn't know if the drugs would work on the other 96 percent of patients with the disease," says Srivastava.

Fortunately, Anna Malashicheva, PhD, from the Russian Academy of Sciences, had collected valve cells from over 20 patients at the time of surgical replacement, and Srivastava struck up a fruitful collaboration with her group to do a "clinical trial in a dish."

"We tested the promising molecules on cells from these 20 patients with aortic valve calcification without known genetic causes," Srivastava adds. "Remarkably, the molecule that seemed most effective in the initial study was able to restore the network in these patients' cells as well."

Once they had identified a promising candidate in cells in a dish for both NOTCH1 and sporadic cases of calcific aortic valve disease, Srivastava and his team did a "pre-clinical trial" in a mouse model of the disease. They wanted to determine whether the drug-like molecule would actually work in a whole, living organ.

The scientists confirmed that the therapeutic candidate could successfully prevent and treat aortic valve disease. In young mice who had not yet developed the disease, the therapy prevented the calcification of the valve. And in mice that already had the disease, the therapy actually halted the disease and, in some cases, led to reversal of the disease. This finding is especially important since most patients aren't diagnosed until calcification has already begun.

"Our strategy to identify gene networkcorrecting therapies that treat the core disease mechanism may represent a compelling path for drug discovery in a range of other human diseases," says Theodoris. "Many therapeutics found in the lab don't translate well to humans or focus only on a specific symptom. We hope our approach can offer a new direction that could increase the likelihood of candidate therapies being effective in patients."

The researchers' strategy relied heavily on technological advancements, including human iPS cells, gene editing, targeted RNA sequencing, network analysis, and machine learning.

"Our study is a really good example of how modern technologies are facilitating the kinds of discoveries that are possible today, but weren't not so long ago," says Srivastava. "Using human iPS cells and gene editing allowed us to create a large number of cells that are relevant to the disease process, while powerful machine learning algorithms helped us identify, in a non-biased fashion, the important genes for distinguishing between healthy and diseased cells."

"By using all the knowledge we gathered over a decade and a half, combined with the latest tools, we were able to find a drug candidate that can be taken to clinical trials," he adds. "Our ultimate goal is always to help patients, so the whole team is very pleased that we found a therapy that could truly improve lives."

About the Research Project

The paper, "Network-based screen in iPSC-derived cells reveals therapeutic candidate for heart valve disease,"was published online by Science on December 10, 2020.

Other authors include Ping Zhou, Lei Liu, Yu Zhang, Tomohiro Nishino, Yu Huang, Sanjeev S. Ranade, Casey A. Gifford, Sheng Ding from Gladstone; Aleksandra Kostina from the Russian Academy of Sciences; and Vladimir Uspensky from the Almazov Federal Medical Research Centre in Russia.

The work was funded by the California Institute of Regenerative Medicine; the National Heart, Lung, and Blood Institute; and the National Center for Research Resources. Gladstone researchers also received support from the Winslow Family, the L.K. Whittier Foundation, The Roddenberry Foundation, the Younger Family Fund, the American Heart Association, several programs and fellowships at UCSF, residency programs from Boston Children's Hospital and the Harvard Medical School, the Uehara Memorial Foundation, and a Howard Hughes Medical Institute Fellowship of the Damon Runyon Cancer Research Foundation.

About Gladstone Institutes

To ensure our work does the greatest good, Gladstone Institutes focuses on conditions with profound medical, economic, and social impactunsolved diseases. Gladstone is an independent, nonprofit life science research organization that uses visionary science and technology to overcome disease.

Media Contact: Julie Langelier | Assistant Director, Communications | [emailprotected] | 415.734.5000

SOURCE Gladstone Institutes

https://gladstone.org

See the rest here:
A Potential Therapy for One of the Leading Causes of Heart Disease - PRNewswire

Creative Medical Technology Holdings files Patent on Induction of Infectious Tolerance by Ex Vivo Reprogrammed Immune Cells Utilizing ImmCelz Cellular…

PHOENIX, Dec. 10, 2020 /PRNewswire/ --Creative Medical Technology Holdings trading under the ticker symbol CELZ announced today its patent filing based on data covering utilization of the Company's ImmCelz product at generating what is termed in the field of immunology as "infectious tolerance."

Using an animal model of rheumatoid arthritis, investigators demonstrated administration of ImmCel protected mice from immunologically mediated joint damage. Importantly, cells from treated mice were able to reverse disease when transferred to arthritic mice. Detailed scientific analysis revealed that ImmCelz administration caused generation of T regulatory cells and tolerogenic dendritic cells. Both of these cell types have previously been described to possess ability to suppress autoimmunity.

"In 2003, Dr. Weiping Min from the University of Western Ontario and myself published a paper describing the Tolerogenic Loop, in which we were able to perform fully mis-matched cardiac transplants without need for long term immune suppression1." Said Dr. Thomas Ichim, Chief Scientific Officer of the Company. "We are extremely enthusiastic to discover that ImmCelz, which is a personalized immunotherapy can induce similar biological processes and in this case suppress autoimmunity."

Creative Medical Technology Holdings possesses numerous issued patents in the area of cellular therapy including patent no. 10,842,815 covering use of T regulatory cells for spinal disc regeneration, patent no. 9,598,673 covering stem cell therapy for disc regeneration, patent no. 10,792,310 covering regeneration of ovaries using endothelial progenitor cells and mesenchymal stem cells, patent no. 8,372,797 covering use of stem cells for erectile dysfunction, and patent no. 7,569,385 licensed from the University of California covering a novel stem cell type.

"Given that our issued intellectual property covers multi-billion dollar markets, it is critical in our development plans to establish scientific mechanisms of action. By understanding how our products work at a cellular and molecular level, we feel we have an advantage when engaging Big Pharma in discussions for licensing/partnering interactions." Said Timothy Warbington, President and CEO of the Company.

The company intends to publish an update on the overall 2020 activities in the coming weeks.

About Creative Medical Technology Holdings

Creative Medical Technology Holdings, Inc. is a commercial stage biotechnology company specializing in stem cell technology in the fields of urology, neurology and orthopedics and trades on the OTC under the ticker symbol CELZ. For further information about the company, please visitwww.creativemedicaltechnology.com.

Forward Looking Statements

OTC Markets has not reviewed and does not accept responsibility for the adequacy or accuracy of this release. This news release may contain forward-looking statements including but not limited to comments regarding the timing and content of upcoming clinical trials and laboratory results, marketing efforts, funding, etc. Forward-looking statements address future events and conditions and, therefore, involve inherent risks and uncertainties. Actual results may differ materially from those currently anticipated in such statements. See the periodic and other reports filed by Creative Medical Technology Holdings, Inc. with the Securities and Exchange Commission and available on the Commission's website atwww.sec.gov.

Timothy Warbington, CEO [emailprotected] CreativeMedicalHealth.com

Creativemedicaltechnology.com http://www.StemSpine.com http://www.Caverstem.com http://www.Femcelz.com

1https://www.jimmunol.org/content/170/3/1304

SOURCE Creative Medical Technology Holdings, Inc.

Home

Excerpt from:
Creative Medical Technology Holdings files Patent on Induction of Infectious Tolerance by Ex Vivo Reprogrammed Immune Cells Utilizing ImmCelz Cellular...

Donor Stem Cell Transplant Improves Survival in Older Patients with Myelodysplastic Syndrome – Cancer Health Treatment News

A new clinical trial offers the most compelling evidence to date that a donor stem cell transplant can improve survival rates for older patients with higher-risk myelodysplastic syndrome (MDS), Dana-Farber Cancer Institute investigators report at the virtual 62nd American Society of Hematology (ASH) Annual Meeting.

Despite being the only current cure for MDS and widely used for younger patients, transplant generally hasnt been offered to older patients because it has not been proven beneficial in that population. The new trial, conducted by the Blood and Marrow Transplant Clinical Trials Network, is likely to change that, according to study leaders. Involving 384 patients at 34 medical centers across the U.S., the trial found that transplantation of hematopoietic stem cells from compatible donors nearly doubled the survival rate of patients age 50-75.

Transplantation has been underutilized, historically, in this patient group, said study senior author Corey Cutler, MD, MPH, FRCPC, of Dana-Farber. Based on our findings all patients should at least be referred to a transplant center so that those who are eligible and have a suitable donor can undergo transplant and have better survival. It is important to refer these patients early so the transplant center can work on finding an optimal donor right from the get-go.

MDS encompasses a group of disorders in which blood-forming cells in the bone marrow become abnormal, resulting in the production of defective blood cells. In about one in three patients, MDS can progress to acute myeloid leukemia, a rapidly growing cancer of bone marrow cells.

Allogeneic hematopoietic stem cell transplantation replaces a patients abnormal blood-forming stem cells with healthy ones from a compatible donor. Because the procedure hadnt been proven to be helpful for older patients, it hasnt been covered by Medicare for people over age 65 unless done as part of an approved study. Medicare approved the design of the trial and is expected to consider the findings when determining future payment policies.

Participants in the new trial were referred to transplant centers, which searched for suitable stem cell donors. The 260 patients who were matched with a donor within 90 days were assigned to receive a stem cell transplant; the other 124 patients received standard supportive care.

Roughly three years after enrolling in the trial, 47.9% of those slated for transplant were alive, compared to 26.6% of those for whom no donor had been found at the 90-day mark. Survival without a recurrence of leukemia was also higher in those assigned to receive a transplant (35.8%) than in those who were not (20.6%). The researchers observed no significant differences among subgroups and no differences in quality of life between the two study arms.

Cutlerpresented findings on this study at the Whats on the Horizon: Practice-Changing Clinical Trials press briefing on Friday, Dec. 4 at 12:30 p.m. EST. Further details were presented during Session 732, Abstract 75, on Saturday, Dec. 5 at 10:30 a.m. EST.

Cutlers disclosures include a consulting or advisory role for Mesoblast, Generon, Medsenic, Jazz, Kadmon, and Incyte.

Complete details on Dana-Farbers activities at ASH are available online at http://www.dana-farber.org/ash.

This press releasewas originally published on December 4, 2020, by Dana-Farber Cancer Institute. It is republished with permission.

Originally posted here:
Donor Stem Cell Transplant Improves Survival in Older Patients with Myelodysplastic Syndrome - Cancer Health Treatment News

Dr. Kansagra: Quadruplet Therapy for Newly Diagnosed Multiple Myeloma and Combination CAR T-Cell Opportunities – DocWire News

Ankit Kansagra, MD, an assistant professor in theDepartment of Internal Medicineat UT Southwestern Medical Center and assistant director of theOutpatient Stem Cell Transplant Program, talks about the potential for changes to combination therapy for multiple myeloma (MM) with the approval of chimeric antigen receptor T-cell agents and the introduction of quadruplet therapy.

In part three of this interview with Dr. Kansagra, available December 15, he discusses newly approved therapies for MM and highlights drugs in the pipeline.

DocWire News: Do you foresee combination therapy for multiple myeloma changing in the next few years, and how would this impact the treatment paradigm and potentially guideline-directed care?

Dr. Kansagra: Lets talk about patients with newly diagnosed multiple myeloma. Right now, we know that for the majority of the country, the standard of care is a combination of three different medicationmay it be bortezomib or [carfilzomib], those are the proteasome inhibitors, lenalidomide, and dexamethasone.

There is a huge debate nationally, or internationally, about adding a fourth agent improves things. There are questions about improving response and the durability of response. Can you get the response, first? And can you sustain that response? Those are two different questions. We know most of the therapies, as of today, including the triplet therapies, are able to get a response; its the durability that is an issue, and can they persist for a longer time or getting the patients into minimal residual disease (MRD), or the lowest disease possible.

Some call it measurable residual disease, some call it minimal residual disease. The point that comes up too is, Are we going to a quadruplet therapy? And the answer to that is yes. I think we are going to a quadruplet therapy, but For who? is going to be a point and For how long? When do you increase the intensity of therapy? When you decrease the intensity of therapy? Would MRD be a guide to deciding how those therapies are done? Those are going to be some of the crucial factors in deciding. So I dont think quadruplet is needed for every single individual with newly diagnosed myeloma. There are subsets who will benefit from it, and there are subsets who probably do not need that aggressive treatment.

For combination with CAR-T cells, I think thats probably a more challenging question. The first question is, scientifically, should we combine something to CAR T-cell therapy to improve its response? As I mentioned earlier, CAR T-cell therapies in lymphoma have shown certainly more impressive responses than what we might have expected in myeloma. Having said that, the diseases are completely different; the biology is completely different. We certainly dont want to compare apples and oranges here. But obviously, there is a challenge in front of us: How are we going to have these CAR T-cells persist to be effective for a longer period of time? Obviously, there has been an ongoing debate and also a lot of clinical trials ongoing, which are thinking of adding medications to the CAR T-cell therapy treatment.

And that may be medications like immunomodulatory drugs or proteasome inhibitors. IMiDs or [lenalidomide] or [pomalidomide], those are certainly key targets here to think of. Even checkpoint inhibitors have been used in some clinical trials, like PD-1 or PD-L1 inhibitors in some clinical trials. I think those are another avenue of interest.

The more important question, to me, is what combination to use rather than what are the potential mechanisms of relapse? Why is CAR-T not working for individual A compared to B compared to C? Is it because the cells are not persisting? Is it because they are losing the BCMA expression? Is it because the CAR-Ts are not expanding at all? And the intervention for each bucket is potentially going to be a different intervention. Some might need a BCMA-targeted ongoing therapy after a CAR T-cell therapy. That might be a strategy in one population, whereas other might need just augmentation with an IMiD. Maybe there is a third, who does not need any intervention at all.

Scientifically speaking, as a physician-scientist, I would think an important thing for us to understand is what are these different categories of patients where we would intervene differently and appropriately. If Im talking to my patient as a physician, as a clinician, sitting in the patients room and thinking of the challenges in the healthcare system, we loved CAR T-cell therapy that we dont have to combine with anything. The initial two years, we loved it, that you get CAR T-cell therapy. My patients tell me, Hey doc, I feel like I dont have myeloma at all.

Thats the comment I get very often from my patients who have received CAR T-cell therapy. The majority of these guys have got stem cell transplant before and have been on maintenance therapies and some ongoing treatment. When it comes to us doing some combination along with CAR T-cell therapy, that is certainly a bummer from them saying, Hey, this was kind of one of those hopes, where we would have done a one-time thing and it would work forever.

From the patients perspective, from a healthcare cost perspective, from a physician sitting in front of them and trying to think what is the easiest in their patient-reported outcome standpoint, Im not too excited about combining things, but I suppose that it is going toward that way to get the best efficacy. So I think rather than saying, Everybody should get a maintenance therapy, like we do for transplant for a big chunk of our patients or pretty much everybody in the United States, I think we will have to think of a much more scientifically-driven answer about who benefits from it.

Watch part 1 of the interview with Dr. Kansagra where he discusses CAR T-cells in development for MM.

More:
Dr. Kansagra: Quadruplet Therapy for Newly Diagnosed Multiple Myeloma and Combination CAR T-Cell Opportunities - DocWire News

Positive Phase 2 Proof-of-Concept Data for Viralym-M and Burden of Disease Data Presented in Oral Presentations at the 62nd American Society of…

CAMBRIDGE, Mass.--(BUSINESS WIRE)--AlloVir (Nasdaq: ALVR), a late clinical-stage cell therapy company, today announced that results from the Phase 2, proof-of-concept CHARMS study demonstrated that an allogeneic, off-the-shelf, multi-virus specific T cell therapy, Viralym-M (ALVR105), achieved a 93% clinical response and was generally well-tolerated in allogeneic hematopoietic stem cell transplantation (allo-HSCT) patients with at least one drug refractory infection. The findings were highlighted during an oral presentation at the 62nd American Society of Hematology Annual Meeting (ASH). In a second oral presentation, a health outcomes analysis showed the high economic and clinical burden of virus-associated hemorrhagic cystitis (V-HC) in patients following allo-HSCT.

There is an urgent need for new therapies that improve the treatment and prevention of viral infections which have significantly impacted immunocompromised patients as well as burdened the healthcare system, said Agustin Melian, MD, Chief Medical Officer and Head of Global Medical Sciences of AlloVir. The data presented at ASH highlight the potential of Viralym-M in treating immunocompromised patients who are at a greater risk of viral infection.

Oral Presentation: Treatment of Severe, Drug-Refractory Viral Infections with Allogeneic, Off-the-Shelf, Multi-virus Specific T Cell Therapy in Patients Following HSCT: Results from a Phase 2 Study Presenter: Bilal Omer, MD, Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX

Efficacy and safety data of Viralym-M in allo-HSCT recipients with at least one treatment-refractory infection (BKV, CMV, AdV, EBV, HHV-6, and/or JCV) were evaluated in the CHARMS study, a Phase 2 proof-of-concept clinical trial. Allo-HSCT patients who had either failed antiviral therapy or were unable to tolerate standard antivirals were enrolled and received a single infusion of Viralym-M. If a partial response was achieved, patients could receive up to four additional doses after four weeks, at two-week intervals.

Ninety-three percent (93%) of the patients achieved a complete (viral load returning to normal range and resolution of clinical signs/symptoms) or partial (a 50% decrease in viral load and/or 50% improvement of clinical signs/symptoms) response by six weeks post-infusion. One hundred percent (100%) of patients who had two or more viral infections (11 of 11) responded to Viralym-M with 19 of the 23 viral infections across these 11 patients responding to treatment. Treatment with Viralym-M was generally well tolerated.

Patients receiving an allogeneic hematopoietic stem cell transplant are at increased risk for multiple viral infections and diseases, which ultimately put them at risk for serious and life-threatening outcomes, said Bilal Omer, MD, Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital and presenting author. Currently there is a lack of effective treatments and those that are available have significant toxicities. Data from the CHARMS study suggest that Viralym-M has the potential to safely and effectively treat the most common viral infections in patients following allogeneic HSCT and may address a critical unmet need in this patient population.

Oral Presentation: Economic and Clinical Burden of Virus-Associated Hemorrhagic Cystitis in Patients Following Allogeneic Hematopoietic Stem Cell Transplantation Presenter: Joseph P. McGuirk, DO, the University of Kansas Cancer Center, Westwood, KS

U.S. claims data were analyzed to compare health care reimbursement, health resource utilization, and clinical outcomes between allo-HSCT patients with virus-associated hemorrhagic cystitis (V-HC) to those without V-HC. The unadjusted mean reimbursement per patient group was $292,401 higher for patients with V-HC compared to patients without V-HC. Adjusted reimbursements were also significantly higher for V-HC patients with and without GVHD compared to patients without V-HC (p<.0001).

V-HC was associated with increased hospital length of stay and additional days in the intensive care unit. Readmission rates also increased for patients with V-HC compared to those without V-HC (p<.0001). The mean overall hospital LOS was prolonged by 27.1 days (p<0.0001), or 55% longer, for allo-HSCT patients with V-HC versus patients without V-HC.

In an adjusted analysis examining time to all-cause mortality, patients with V-HC had a 70% higher risk of mortality, even after adjusting for presence of GVHD as well as other baseline factors. V-HC was also associated with increased incidence for renal impairment in the follow-up period in patients with or without GVHD.

Viral infections or disease including the viruses associated with hemorrhagic cystitis are a major complication in patients who have undergone allogeneic hematopoietic stem cell transplantation, said Joseph P. McGuirk, DO, the University of Kansas Cancer Center and presenting author. These data show that virus associated hemorrhagic cystitis has a significant impact on patient outcomes including a greater risk of death and new renal impairment, while also placing a substantial burden on our healthcare system, highlighting the unmet clinical need for effective strategies to prevent and/or treat virus-associated hemorrhagic cystitis in allo-HSCT recipients.

Viral Infections in Immunocompromised Patients

In healthy individuals, virus-specific T-cells (VSTs) from the bodys natural defense system provide protection against numerous disease-causing viruses. However, in patients with a weakened immune system these viruses may be uncontrolled. Viral diseases are common and can cause potentially devastating and life-threatening consequences in immunocompromised patients. For example, up to 90% of patients will reactivate at least one virus following an allogeneic stem cell transplant and two-thirds of these patients reactivate more than one virus, resulting in significant and prolonged morbidity, hospitalization, and premature death. Typically, when viruses infect immunocompromised patients, standard antiviral treatment does not address the underlying problem of a weakened immune system and therefore many patients suffer with life-threatening outcomes such as multi-organ damage and failure, and even death.

Viralym-M (ALVR105)

AlloVirs lead product Viralym-M (ALVR105) is in late-stage clinical development as an allogeneic, off-the-shelf, multi-virus specific T-cell therapy targeting six common viral pathogens in immunocompromised individuals: BK virus, cytomegalovirus, adenovirus, Epstein-Barr virus, human herpesvirus 6, and JC virus. The company plans to initiate a Phase 3, multicenter, double-blind, placebo-controlled study to assess the efficacy and safety of Viralym-M for the treatment of patients with virus-associated hemorrhagic cystitis (V-HC) following allo-HSCT before year end. A proof-of-concept clinical trial targeting the prevention of BKV, CMV, AdV, EBV, HHV-6, and JCV in patients following allo-HSCT is also expected to initiate by the end of 2020. Viralym-M has received Regenerative Medicine Advanced Therapy (RMAT) designation from the U.S. Food and Drug Administration (FDA), as well as PRIority MEdicines (PRIME) and Orphan Drug Designations (ODD) from the European Medicines Agency.

About AlloVir

AlloVir is a leading late clinical-stage cell therapy company with a focus on restoring natural immunity against life-threatening viral diseases in patients with weakened immune systems. The companys innovative and proprietary technology platforms leverage off-the-shelf, allogeneic, multi-virus specific T cells targeting devastating viruses for patients with T cell deficiencies who are at risk from the life-threatening consequences of viral diseases. AlloVirs technology and manufacturing process enables the potential for the treatment and prevention of a spectrum of devastating viruses with each single allogeneic cell therapy. The company is advancing multiple mid- and late-stage clinical trials across its product portfolio. For more information visit http://www.allovir.com.

Forward-Looking Statements

This press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995, as amended, including, without limitation, statements regarding AlloVirs development and regulatory status of our product candidates and its strategy, business plans and focus. The words may, will, could, would, should, expect, plan, anticipate, intend, believe, estimate, predict, project, potential, continue, target and similar expressions are intended to identify forward-looking statements, although not all forward-looking statements contain these identifying words. Any forward-looking statements in this press release are based on managements current expectations and beliefs and are subject to a number of risks, uncertainties and important factors that may cause actual events or results to differ materially from those expressed or implied by any forward-looking statements contained in this press release, including, without limitation, those related to AlloVirs financial results, the timing for completion of AlloVirs clinical trials of its product candidates, whether and when, if at all, AlloVirs product candidates will receive approval from the U.S. Food and Drug Administration, or FDA, or other foreign regulatory authorities, competition from other biopharmaceutical companies, and other risks identified in AlloVirs SEC filings. AlloVir cautions you not to place undue reliance on any forward-looking statements, which speak only as of the date they are made. AlloVir disclaims any obligation to publicly update or revise any such statements to reflect any change in expectations or in events, conditions or circumstances on which any such statements may be based, or that may affect the likelihood that actual results will differ from those set forth in the forward-looking statements. Any forward-looking statements contained in this press release represent AlloVirs views only as of the date hereof and should not be relied upon as representing its views as of any subsequent date.

Original post:
Positive Phase 2 Proof-of-Concept Data for Viralym-M and Burden of Disease Data Presented in Oral Presentations at the 62nd American Society of...

Silicon Therapeutics Announces Members of Scientific Advisory Board – Business Wire

BOSTON--(BUSINESS WIRE)--Silicon Therapeutics, a privately-held, integrated therapeutics company with a pioneering drug discovery platform based on physics-driven molecular simulations, today announced the members of the Silicon Therapeutics scientific advisory board (SAB), which include Dr. Elliot L. Chaikof, Dr. Timothy P. Heffernan, Dr. Sun Hur, Dr. Pasi A. Jnne and Dr. Lijun Sun.

The Silicon Therapeutics SAB is comprised of experts in biophysics, medicinal chemistry, translational medicine and research and development (R&D) in oncology and immunology. These experts will serve as a strategic resource for Silicon Therapeutics to provide scientific review and high-level advice about the companys drug discovery strategy as it continues to advance the companys therapeutic pipeline using its proprietary simulation platform.

We are honored to officially welcome Drs. Chaikof, Heffernan, Hur, Jnne and Sun as members of our SAB, said Christopher Winter, Ph.D., chief of research and development. Each members expertise, insights and direction will provide a tremendous knowledge base that will help inform our approach regarding research and clinical development activities, which will be invaluable as we head into the next phase of our growth advancing our in-house research programs developed utilizing our proprietary drug discovery platform.

The SAB will be involved in strategic discussions related to targets, research and pre-clinical development, as well as the next generation, first-in-class therapeutic pipeline.

Silicon Therapeutics is focused on the discovery and development of first-in-class small molecules targeting key drivers of disease in cancer and inflammation that have proven difficult to treat with prior approaches and thus previously considered undruggable. The companys unique discovery platform is fully integrated with Silicon Therapeutics internal laboratories using cutting edge experimental capabilities in biophysics, biology and chemistry.

ABOUT THE SILICON THERAPEUTICS SCIENTIFIC ADVISORY BOARD

Elliot L. Chaikof, M.D., Ph.D. is a co-founder of Silicon Therapeutics and chair of the Roberta and Stephen R. Weiner department of surgery and surgeon-in-chief at the Beth Israel Deaconess Medical Center (BIDMC), as well as the Johnson & Johnson professor of surgery at Harvard Medical School in Boston. Dr. Chaikof is a member of the Wyss Institute of Biologically Inspired Engineering of Harvard University and the Harvard Stem Cell Institute, also in Boston.

Dr. Chaikof earned a bachelor of arts degree and medical doctor degree from Johns Hopkins University in Baltimore and a doctor of philosophy degree in chemical engineering from the Massachusetts Institute of Technology in Boston, where he focused on the design of artificial organs. Dr. Chaikof completed his training in general surgery at the Massachusetts General Hospital in Boston and in vascular surgery at Emory University School of Medicine in Atlanta.

Timothy P. Heffernan, Ph.D. is head of oncology research within the division of therapeutics discovery and development at The University of Texas MD Anderson Cancer Center in Houston. Dr. Heffernan also serves as executive director for the Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION) platform, a translational research unit focused on accelerating the pre-clinical evaluation of novel drugs to inform innovative clinical trials.

Dr. Heffernan earned a doctor of philosophy degree in cell and molecular pathology from the University of North Carolina at Chapel Hill in Chapel Hill, N.C. and performed his postdoctoral training at the Dana-Farber Cancer Institute and Harvard Medical School in Boston.

Sun Hur, Ph.D. is an Oscar M. Schloss professor in the department of biological chemistry and molecular pharmacology in the department of pediatrics at Harvard Medical School in Boston.

Dr. Hur earned a bachelor of science degree in physics from Ewha Womans University in Seoul, South Korea and a doctor of philosophy degree in physical chemistry with Dr. Thomas C. Bruice at the University of California, Santa Barbara. Dr. Hur did her post-doctoral work in x-ray crystallography with Dr. Robert M. Stroud at the University of California, San Francisco. She joined Harvard Medical School in 2008 as an assistant professor and became affiliated with Boston Childrens Hospital in 2010.

Pasi A. Jnne, M.D., Ph.D. is a thoracic medical oncologist at the Dana-Farber Cancer Institute, a professor at Harvard Medical School, director of the Lowe Center for Thoracic Oncology and director of the Belfer Center for Applied Cancer Science in Boston.

Dr. Jnne earned a medical doctor degree and doctor of philosophy degree at the University of Pennsylvania in Philadelphia. He completed postgraduate training in internal medicine at Brigham and Womens Hospital and in medical oncology at Dana-Farber Cancer Institute in Boston.

Lijun Sun, Ph.D. is a co-founder of Silicon Therapeutics and an associate professor of surgery at Harvard Medical School, Beth Israel Deaconess Medical Center in Boston.

Dr. Sun earned a doctor of philosophy degree in organic chemistry from Emory University in Atlanta and completed postdoctoral training in biomaterial research at Emory School of Medicine in Atlanta.

ABOUT SILICON THERAPEUTICS

Silicon Therapeutics is a privately held, fully integrated drug design and development company focused on small molecule therapeutics. The Silicon Therapeutics proprietary physics-driven drug design platform combines quantum physics, statistical thermodynamics, molecular simulations, a dedicated HPC super-computing cluster, purpose-built software, in-house laboratories and clinical development capabilities. The platform was built from the ground up to address difficult targets using physics-based simulations and experiments to pioneer a new path for drug design with the prime goal of delivering novel medicines to improve the lives of patients.

Silicon Therapeutics is currently the only company that owns the entire spectrum of proprietary physics-driven drug discovery from chip-to-clinic. The companys lead program is a highly differentiated small molecule Stimulator of Interferon Genes (STING) agonist for the treatment of cancer, which entered the clinic in November 2020. The companys headquarters are located in Boston. To learn more about Silicon Therapeutics, please visit our website at http://www.silicontx.com or follow us on LinkedIn and Twitter.

More:
Silicon Therapeutics Announces Members of Scientific Advisory Board - Business Wire

Israeli Neurogenesis’ NG-01 slows progressive MS by up to 90% in phase II study – BioWorld Online

Israeli cell therapy specialist Neurogenesis Ltd., said new phase II data has shown that treatment with its autologous cell therapy candidate NG-01 in progressive multiple sclerosis (MS) patients led to an 80% to 90% reduction in disease progression at 12 months compared to a pretreatment period and a 90% reduction in relapses compared to placebo-treated patients.

The results are highly significant in all parameters and show much more efficacy than any other treatment in progressive MS, Dimitrios Karussis, lead principal investigator and director of the MS Center at Hadassah Medical Center in Jerusalem, told BioWorld.

Karussis explained that there are currently very effective medications for stopping relapses and slowing the progression of relapsing remitting MS, but not very successful ones for progressive disease. The only registered treatment for primary progressive MS, Roche Holding AGs Ocrevus (ocrelizumab), slows down the progression rate by 20% to 25%, he said. For secondary progressive MS, Novartis AG's Mayzent (siponimod) also slows progression by about 20%, he added.

To determine whether NG-01 might one day prove more effective, the phase II study enrolled 48 people with progressive MS who were randomized into three groups and assigned to receive either an intrathecal or I.V. dose of NG-01 or a placebo injection. The study lasted for 14 months and met all of its primary endpoints.

Researchers said no serious treatment-related safety issues were detected. Only 6.7% and 9.7% of patients experienced disease progression in the intrathecal and I.V. NG-01 treatment groups, respectively, compared to 41.9% in the placebo group (p=0.0003 and p=0.0008). Also, 58% of the patients treated intrathecally with NG-01 did not show any evidence of disease activity during the entire the treatment period, compared to 9.7% in the placebo treated group (p<0.0001).

Trial participants treated with NG-01 demonstrated a significant improvement in walking ability, as measured by 25-foot walking time (P=0.0017), the company said. Investigators also found that intrathecal administration of NG-01 was also more efficacious than I.V. delivery in several key parameters of the disease, such as relapse rate (89% decrease in the relapse rate), functional MRI, monthly changes of the MRI T2 lesion load and the 9-hole peg test vs. the placebo-treated group.

The strengths of our trial include the inclusion of patients with active progressive multiple sclerosis, for which existing immunotherapies are usually ineffective, the double-blind design making this the first randomized controlled trial comparing intrathecal versus intravenous methods of NG-01 administration and single versus repeated treatment; and the robust, though short-term (6-12 months), clinical benefits observed in several disease activity parameters, including newer biomarkers, such as functional MRI-network connectivity, OCT, and cognitive testing, Karussis told BioWorld.

Separately, he said this trial provides encouraging results and suggests a potential for a new approach that may not only slow down the progression of the disease but even induce improvement and promote repair mechanisms in progressive MS.

Following the encouraging phase II data, Neurogenesis CEO Tal Gilat told BioWorld the next step is to start a multicenter phase IIb in the U.S., Europe and Israel, which will focus on improvement and repair in progressive MS patients, he said. The company has already had a pre-IND meeting with the FDA.

NG-01 is Neurogenesis lead product. Besides being tested in progressive MS patients, it was also tested in two phase IIa trials in amyotrophic lateral sclerosis.

Given our positive outcomes in two open-label trials in ALS with long follow-up periods, we are now considering what would be the best route to clinically advance NG-01 also for the ALS indication, Gilat said.

The Jerusalem-based biotech focuses on developing cell therapies for neurodegenerative diseases with an approach for sustained delivery of high levels of remyelinating growth factors using the patients own stem cells.

Neurogenesis licensed NG-01 from Hadasit, the technology transfer company of Hadassah Medical Center, to advance the cell therapy candidate.

The biotech used its Neuralyzed Cell platform to identify, culture and enhance a subpopulation of bone marrow cells towards remyelinating cells that also possess neurotrophic immunomodulatory and neuroprotective properties. The NG-01 cell population is injected directly into the central nervous system through the cerebrospinal fluid, where the cells home-in on a damaged area, take up residence and produce large amounts of neurotrophic factors.

Neurogenesis helped optimized the cell manufacturing process to become highly efficient and scalable, Gilat said. By manufacturing the required treatments from one bone marrow extraction, storing the vials in cryopreservation, and shipping them to the hospital only when required, we enable high consistency of quality across injections, while lowering manufacturing costs as well as enabling treatment schedule flexibility.

Read the original:
Israeli Neurogenesis' NG-01 slows progressive MS by up to 90% in phase II study - BioWorld Online