Category Archives: Stem Cell Treatment


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...

Hematologist/Stem Cell Biologist to Direct Hematology and Cellular Therapy at Cedars-Sinai – Newswise

Newswise LOS ANGELES (Dec. 1, 2020) -- Internationally recognized hematologist John P. Chute, MD, has been selected to direct the Division of Hematology and Cellular Therapy in the Department of Medicine at Cedars-Sinai Cancer. The physician-scientist also will serve as director of the Center for Myelodysplastic Diseases Research and associate director of the Board of Governors Regenerative Medicine Institute in the Department of Biomedical Sciences. Chute assumed his new post Nov. 23.

The selection of Chute, following a national search, reflects the importance of his pioneering research in blood-forming stem cells called hematopoietic stem cells, which can self-renew and generate all cell types found in the blood and immune system. Over the past decade, Chute's lab has discovered several growth factors produced by the cells that line the walls of blood vessels; they play a critical role in blood-forming stem cell regeneration.

"Dr. Chute is an exceptional addition to our faculty," saidDanTheodorescu, MD, PhD, director ofCedars-Sinai Cancer. "His international reputation as a physician-scientist who has made major contributions to stem cell and hematopoietic cell biologywill greatly contribute to positioning the newly created Division of Hematology and Cell Therapy as one of the best in the nation, while providing Cedars-Sinai Cancer patients with exciting new options for the treatment of blood malignancies."

In addition to his hematopoietic stem cell research, Chute said he looks forward to expanding Cedars-Sinai's CAR T-cell research and therapy. He describes the immune-boosting therapeutic as "transformative" for patients with advanced non-Hodgkinlymphoma,childhood acute lymphoblastic leukemiaand potentially several additional blood cancers.

CAR T-cell therapy is a type of immunotherapy in which patients' own immune cells, called T cells, are collected from their blood, and then an artificial receptor chimeric antigen receptor, or CAR is added to the cells' surface. The receptor enables the modified cells to specifically eradicate cancer cells. The cells are infused back into a patient's body intravenously, where they multiply and attack tumor cells.

"CAR T therapy has become an important treatment option for so many patients with advanced cancer who had no options before," Chute said. "That's what makes CAR T therapy so exciting."

Chute joins Cedars-Sinai from the David Geffen School of Medicine at the UCLA, where he was a professor of Medicine and Radiation Oncology in the Division of Hematology/Oncology and an investigator in the Broad Stem Cell Research Center.

Chute earned his medical degree at Georgetown University. He completed his residency in internal medicine and fellowship in Hematology/Oncology at the National Naval Medical Center. He completed his research training at the National Cancer Institute and the Naval Medical Research Institute.

"I'm excited to join the Cedars-Sinai Cancer faculty because of the opportunity to collaborate with the world-class scientists and top-tier physicians at the cancer center," Chute said. "Cedars-Sinai has always been a leading medical center and is deeply committed to basic and translational research, while also growing the hematology and cellular therapy specialties. I'm eager to play a leading role in that growth."

Clickhereto read more from the Cedars-Sinai Newsroom.

Continued here:
Hematologist/Stem Cell Biologist to Direct Hematology and Cellular Therapy at Cedars-Sinai - Newswise

Boston Celtics Kemba Walker will miss time after stem cell injection in left knee, will be re-evaluated in J – MassLive.com

Boston Celtics guard Kemba Walker will miss time at the start of the season after receiving a stem-cell injection in his left knee, the team announced Tuesday morning.

Per the Celtics, Walker consulted with multiple specialists before settling on a treatment in early October. His timeline for a return was 12 weeks, and he will be re-evaluated in early January. The NBA season is scheduled to open on Dec. 22, so Walker will miss time and then could be limited afterward as the Celtics try to tend to his knee as best they can.

Walker struggled with knee issues all year, playing in just 56 of Bostons regular-season games. He appeared limited at times when the season resumed in the Disney World bubble as well, despite a four-month layoff.

Walker, who is 30, signed a four-year, $140 million contract with the Celtics in 2019.

Per the Celtics, second-year wing Romeo Langford will also miss time. He had a procedure to repair a torn scapholunate ligament in his right wrist in September and was expected to miss four-to-five months. Per the Celtics, his recovery is proceeding on schedule.

Tristan Thompson also suffered a minor hamstring strain and will for the first week of training camp.

Related content

Boston Celtics announce official signings of Tristan Thompson, Jeff Teague

Boston Celtics rumors: Evan Turner will join team on the bench as assistant coach (report)

Gordon Hayward trade: Who Boston Celtics can target, what a traded player exception is, & how to use it

Visit link:
Boston Celtics Kemba Walker will miss time after stem cell injection in left knee, will be re-evaluated in J - MassLive.com

Stem cell therapy in coronavirus disease 2019: current evidence and future potential – DocWire News

This article was originally published here

Cytotherapy. 2020 Nov 9:S1465-3249(20)30932-4. doi: 10.1016/j.jcyt.2020.11.001. Online ahead of print.

ABSTRACT

The end of 2019 saw the beginning of the coronavirus disease 2019 (COVID-19) pandemic that soared in 2020, affecting 215 countries worldwide, with no signs of abating. In an effort to contain the spread of the disease and treat the infected, researchers are racing against several odds to find an effective solution. The unavailability of timely and affordable or definitive treatment has caused significant morbidity and mortality. Acute respiratory distress syndrome (ARDS) caused by an unregulated host inflammatory response toward the viral infection, followed by multi-organ dysfunction or failure, is one of the primary causes of death in severe cases of COVID-19 infection. Currently, empirical management of respiratory and hematological manifestations along with anti-viral agents is being used to treat the infection. The quest is on for both a vaccine and a more definitive management protocol to curtail the spread. Researchers and clinicians are also exploring the possibility of using cell therapy for severe cases of COVID-19 with ARDS. Mesenchymal stromal cells are known to have immunomodulatory properties and have previously been used to treat viral infections. This review explores the potential of mesenchymal stromal cells as cell therapy for ARDS.

PMID:33257213 | DOI:10.1016/j.jcyt.2020.11.001

See the rest here:
Stem cell therapy in coronavirus disease 2019: current evidence and future potential - DocWire News

Hematologist Discusses the Impact a Myeloma CAR T-Cell Approval Would Have on the Treatment Landscape – 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, discusses chimeric antigen receptor T-cell agents in the pipeline for multiple myeloma (MM) and how these therapies may impact the treatment landscape pending future approvals.

In part two of this interview with Dr. Kansagra, available December X, he discusses potential new combination therapy options for MM.

DocWire News: Dr. Kansagra, can you discuss some of the CAR T-cell therapies in development for multiple myeloma, including their targets, clinical trial data that weve seen, and your expectations for any future FDA approvals?

Dr. Kansagra: In multiple myeloma, a few of the CAR T-cell therapy targets, which in the most developments, have been the BCMA-targeted CAR T-cell therapies. Those have been most exciting because they have made it to the phase I to phase II trials, especially the registrational studies from Celgene or Bluebird, BMS, the bb2121 compound or the Janssen compound 4538, being farthest out in the clinical development for CAR T-cell therapy. There are certainly a few other CAR T-cell therapies for multiple myeloma, which have grown, and theyre probably in the earlier development of therapy. An example being the CD38 CAR T-cell therapy, the SLAMF CAR T-cell therapy, and GPR5CD CAR T-cell therapy. Those are the three different targets which are being evaluated as T-cell targets.

DocWire News: How do you see the approval of these CAR T-cell therapy impacting the treatment landscape for multiple myeloma?

Dr. Kansagra: I think its going to be a huge improvement in our momentum of our treatment options. We have already seen cell therapy in myeloma have impressive results in terms of the response rates. I think the first important step is you have these patients who have got six or seven different lines of treatment, and now they are getting a novel product or a novel mechanism of action and also novel target and seeing an impressive response rate. That was amazing. Thats step number one.

Step number two is, as we have got further into the clinical development of CAR T-cell therapy, we have seen the safety of these products because that is extremely important that our products are safer.

Then the third thing which we have seen is that long-term follow-ups are not there, but what we have started seeing is that our responses, which could last up to a year or a year and a half for the population, where we would have usually seen maybe barely a response in a matter of months.

I think those are exciting times for our patients with multiple myeloma, where they have failed a lot of therapies. I think the more exciting times are going to come when we will start seeing these CAR T-cell therapies, potentially even in earlier lines of treatment options, where they could use maybe as a second-line treatment or as a first-line treatment after stem cell transplant or in lieu of stem cell transplant, maybe we can have deeper and longer remission rates.

DocWire News: With some of these agents potentially coming to market, do you foresee any challenges, either associated with adverse events or the ability to make these treatments widely available to patients?

Dr. Kansagra: Access to care is certainly near and dear to me, and thinking about those challenges is extremely, extremely important. I think were going to probably face challenges in a lot of different ways.

The first thing is, obviously, how can we get our patients to the centers who are giving CAR T-cell therapy? How are we going to bring them? We know from our autologous stem cell transplant over the last three to four decades, that still not every eligible transplant patient is referred to a transplant center, for whatever reasons. There are multiple reasons; there are socioeconomic reasons; there are distance reasons. But a lot of them are fixable reasons. There are some which are unfixable, but there are some fixable. I think the first and the foremost important thing is going to be to get our patients to a place who is delivering CAR T-cell therapy. Thats the challenge number one.

Challenge number two is, once they are in there, making sure that they are able to get that thing. So it means theyre not coming too late in their game, so trying to make sure theyre referred in earlier points, so that processes in place, that insurance approval has got started, if we need to work on the sociodemographic issues, how are they going to stay in a particular area? What is the social help, what is the family help theyre going to need? If they had referred earlier on, thats another, I call it, bottleneck that we need to think of that. Thats where we need to act on it.

The hard thing is obviously the cost. We dont know what is going to be the cost of the myeloma CAR T-cell therapy, or what is the price of those things. We can certainly estimate that its not going to be as cheap given the three CAR-Ts, which are not FDA-approved. I think its going to be expensive. You will have to think of the cost of care model of how we are going to work with this.

Last but not least of the challenges are the CAR-T itself. These are in the logistical challenge bucket. Then there are the challenges in the CAR-T landscape or the product itself. We still know that these are second-generation CAR T-cell therapies. They dont work for everybody. They have a high response rates, but they dont last that long. We hope to see longer remissions. An example I give, in comparison to large-cell lymphoma, we had 50% of the people who plateaued out, now coming up to about three years. In myeloma, we havent obviously made it to three years since the CAR T-cell therapy have started, but we do worry that there is a tail end of the curve that people are already relapsing to it. Obviously, that goes to the product itself or the construct itself, which needs to be developed in multiple different ways. I think of them as two major challenges ahead of us.

Originally posted here:
Hematologist Discusses the Impact a Myeloma CAR T-Cell Approval Would Have on the Treatment Landscape - DocWire News

Sphingosine 1-phosphate Receptor Modulator ONO-4641 Regulates Trafficking of T Lymphocytes and Hematopoietic Stem Cells and Alleviates Immune-Mediated…

This article was originally published here

J Pharmacol Exp Ther. 2020 Nov 30:JPET-AR-2020-000277. doi: 10.1124/jpet.120.000277. Online ahead of print.

ABSTRACT

ONO-4641 is a second-generation sphingosine 1-phosphate (S1P) receptor modulator that exhibits selectivity for S1P receptors 1 and 5. Treatment with ONO-4641 leads to a reduction in magnetic resonance imaging disease measures in patients with relapsing-remitting multiple sclerosis. The objective of this study was to explore the potential impact of ONO-4641 treatment based on its immunomodulatory effects. Severe aplastic anemia is a bone marrow (BM) failure disease, typically caused by aberrant immune destruction of blood progenitors. Although the T helper type-1-mediated pathology is well described for aplastic anemia, the molecular mechanisms driving disease progression remain undefined. We evaluated the efficacy of ONO-4641 in a mouse model of aplastic anemia. ONO-4641 reduced the severity of BM failure in a dose-dependent manner, resulting in higher blood and BM cell counts. By evaluating the mode of action, we found that ONO-4641 inhibited the infiltration of donor-derived T lymphocytes to the BM. ONO-4641 also induced the accumulation of hematopoietic stem cells in the BM of mice. These observations indicate, for the first time, that S1P receptor modulators demonstrate efficacy in the mouse model of aplastic anemia and suggest that treatment with ONO-4641 might delay the progression of aplastic anemia. Significance Statement ONO-4641 is a second-generation sphingosine 1-phosphate (S1P) receptor modulator selective for S1P receptors 1 and 5. In this study, we demonstrated that ONO-4641 regulates the trafficking of T lymphocytes along with hematopoietic stem and progenitor cells leading to alleviation of pancytopenia and destruction of bone marrow in a bone marrow failure-induced mouse model mimicking human aplastic anemia.

PMID:33257316 | DOI:10.1124/jpet.120.000277

See more here:
Sphingosine 1-phosphate Receptor Modulator ONO-4641 Regulates Trafficking of T Lymphocytes and Hematopoietic Stem Cells and Alleviates Immune-Mediated...