Biocardia Announces FDA-Recommended Modifications to Primary Endpoint for Cardiamp Cell Therapy Heart Failure Trial to Support Marketing Approval -…

SAN CARLOS, Calif., April 30, 2020 (GLOBE NEWSWIRE) -- BioCardia, Inc.[Nasdaq: BCDA], a leader in the development of comprehensive solutions for cardiovascular regenerative therapies, today reported that it has accepted and implemented FDA-recommended modifications to the primary endpoint for the CardiAMP Cell Therapy Heart Failure Trial and associated statistical analysis plan. The Agency proposed these modifications to support the potential for marketing approval for the therapy based on the currently enrolling pivotal trial.

The primary endpoint going forward will be an outcomes composite score based on a three-tiered Finkelstein-Schoenfeld (FS) hierarchical analysis, an established outcomes design that has been used in other leading heart failure programs. The FS procedure is a ranked analysis that first compares each subject to each other subject for occurrence of first-tier events (time to death, in this case) and then compares patient outcomes for subsequent tiers. The tiers, starting with the most serious events, would be (1) all-cause death, including cardiac death equivalents such as heart transplant or left ventricular assist device placement, ordered by time to event; (2) non-fatal Major Adverse Coronary and Cerebrovascular Events (MACCE), excluding those deemed procedure-related occurring within the first seven days post-procedure (heart failure hospitalization, stroke or myocardial infarction), ordered by time to event, and (3) change from baseline in Six Minute Walk Distance at 12 months.

BioCardia CEO Peter Altman, PhD, said, The only major modification to our previous endpoint is that patients who experience a MACCE event and recover will not be factored into the benefit seen in the third-tier measure of Six Minute Walk Distance at one-year follow-up. Since we saw no incidence of MACCE at one year among treated patients in our Phase II trial, our probability of achieving a positive result in the primary endpoint in the ongoing pivotal CardiAMP Heart Failure trial remains unchanged, at greater than 95 percent. Because the FS composite outcomes endpoint is already established for heart failure trials, we expect it to significantly enhance understanding and confidence in our trial results among physicians and payers alike.

The CardiAMP Heart Failure Trial is studying CardiAMP cell therapy, an autologous bone marrow-derived mononuclear cell formulation designed to stimulate the bodys natural healing response in treating heart failure which develops after a heart attack. The trial is evaluating the cell therapys ability to improve patient survival, exercise capacity and quality of life, as well as its safety. The CardiAMP Heart Failure Trial is the first multicenter clinical trial of an autologous cell therapy to prospectively select patients based on cell potency to maximize the probability of patient benefit.

The ongoing multi-center, double-blinded, randomized (3:2), controlled pivotal CardiAMP Heart Failure Trial is expected to enroll 260 patients at up to 40 centers nationwide. The national co-principal investigators are Amish Raval, MD, of the University of Wisconsin and Carl Pepine, MD, of the University of Florida, Gainesville. In March 2020, the Data Safety Monitoring Board indicated there were no safety concerns with the CardiAMP study results and recommended that the trial continue, as planned.To date, 74 patients have been enrolled at 25 active centers. The trial is sponsored, in part, by the Maryland Stem Cell Research Foundation and has reimbursement from the Centers for Medicare and Medicaid Services (CMS).

For additional resources and to learn more about the CardiAMP Heart Failure Trial, visit http://www.biocardia.com.

About BioCardiaBioCardia, Inc., headquartered in San Carlos, California, is developing regenerative biologic therapies to treat cardiovascular disease. CardiAMP and CardiALLO cell therapies are the Companys biotherapeutic product candidates in clinical development. The Company's approved products include the Helix transendocardial delivery system and its steerable guide and sheath catheter portfolio. BioCardia also partners with other biotherapeutic companies to provide its Helix System and clinical support to their programs studying therapies for the treatment of heart failure, chronic myocardial ischemia and acute myocardial infarction.

Forward Looking Statements This press release contains forward-looking statements that are subject to many risks and uncertainties. Forward-looking statements include, among other things, the intended outcomes of our trials, the availability of data from our clinical trials, filings with the FDA, FDA product clearances, the efficacy and safety of our products and therapies and statements regarding our intentions, beliefs, projections, outlook, analyses or current expectations. Such risks and uncertainties include, among others, the inherent uncertainties associated with developing new products or technologies, regulatory approvals, unexpected expenditures, the ability to raise the additional funding needed to continue to pursue BioCardias business and product development plans and overall market conditions. These forward-looking statements are made as of the date of this press release, and BioCardia assumes no obligation to update the forward-looking statements.

We may use terms such as believes, estimates, anticipates, expects, plans, intends, may, could, might, will, should, approximately or other words that convey the uncertainty of future events or outcomes to identify these forward-looking statements. Although we believe that we have a reasonable basis for each forward-looking statement contained herein, we caution you that forward-looking statements are not guarantees of future performance and that our actual results may differ materially from the forward-looking statements contained in this press release. As a result of these factors, we cannot assure you that the forward-looking statements in this press release will prove to be accurate. Additional factors that could materially affect actual results can be found in BioCardias Form 10-K filed with the Securities and Exchange Commission on April 9, 2020, under the caption titled Risk Factors. BioCardia expressly disclaims any intent or obligation to update these forward-looking statements, except as required by law.

Media Contact: Michelle McAdam, Chronic Communications, Inc.Email:michelle@chronic-comm.comPhone: 310-902-1274

Investor Contact: David McClung, Chief Financial OfficerEmail:investors@BioCardia.comPhone: 650-226-0120

Read more here:
Biocardia Announces FDA-Recommended Modifications to Primary Endpoint for Cardiamp Cell Therapy Heart Failure Trial to Support Marketing Approval -...

Novel Bispecific CD19/CD22 CAR-T Therapy Deemed Tolerable in Relapsed/Refractory ALL – Cancer Therapy Advisor

A novel,bispecific CD19/CD22 chimeric antigen receptor T-cell (CAR-T) therapy wastolerable and resulted in responses among patients with acute lymphoblasticleukemia (ALL), according to results from a phase 1 trial presented at the AmericanAssociation for Cancer Research (AACR) Virtual Annual Meeting I 2020.

Thenovel CAR-T therapy was developed with the hypothesis that dual antigen-targetingstrategies may prevent antigen negative escape, Haneen Shalabi, DO, of theNational Cancer Institute and lead author and presenter of the study, said.

Thephase 1, dose-escalation study treated 13 young patients with ALL with theCD19/CD22 CAR-T therapy at 3 different dose levels, including 3 x 105,1 x 106, and 3 x 106. The bispecific construct containedFMC63 (CD19 scFv) linked with m971 (CD22 scFv) and a 4-1 BB costimulatorydomain.

Patientsunderwent lymphodepletion with fludarabine plus cyclophosphamide prior to theirCAR-T infusion. The primary endpoints were safety and toxicity, and thesecondary endpoints were efficacy, chimeric antigen receptor (CAR) expansion,and CAR persistence.

Atbaseline, the median age was 19.6 (range, 5.4-28.5). Patients had receivedprevious treatments, including hematopoietic stem cell transplant (54%),CD19-targeted therapy (69%), prior CD19 CAR T cell therapy (38.4%),blinatumomab (61.5%), CD22-targeted therapy (38.4%), inotuzumab (30.7%), andCD22 CAR-T therapy (15.4%). Extramedullary disease was present in 46.2% ofpatients.

CAR Tcells were well tolerated and toxicities were reversible in all patients, DrShalabi said.

Cytokinerelease syndrome (CRS) developed in 46% of patients, 15.4% of which was grade 3or higher. Both patients who developed grade 3 or higher CRS had received the 1x 106 dose level of the CD19/CD22 CAR-T product and both requiredtreatment with tocilizumab. One patient developed neurotoxicity, and hadreceived the 3 x 106 dose level.

Of the12 patients evaluable for efficacy, a complete response (CR) was achieved by42% (5) of patients, including all patients who received the 1 x 106 or 3 x 106 dose levelsof the CD19/CD22 CAR-T therapy. There were 2 nonresponders.

Two patientswho received 1 x 106 CAR-T and all patients who received the 3 x 106dose level were negative for minimal residual disease (MRD), with the remainingCRs demonstrating bone marrow clearance. Four of the 5 patients who were MRDnegative were also naive to CAR-T therapy.

Of the 5patients who achieved a CR, 2 relapsed with CD19-positive/CD22-positive diseaseand 3 remained in remission at a median 7 months after CAR T cell infusion.

Severalpatients, however, who were MRD negative in the bone marrow did not achieve CRin their extramedullary disease. Dr Shalabi said that these discrepant resultsbetween marrow and extramedullary disease suggests potentially limited CAR-Ttrafficking to sites of extramedullary disease. She suggested that treatmentat higher dose levels may be needed to overcome this limitation.

CAR T-cellexpansion occurred in all patents who responded, with a median peak inperipheral blood of 7%. At day 28, there were 1.3% CAR T cells in the bonemarrow. The persistence of the CAR T cells in peripheral blood was a median of45.6 days, as measured by flow cytometry.

Dr Shalabi concluded that this early experience with bispecific CD19/CD22 CAR T cells demonstrates clinical activity with reversible CRS and limited neurotoxicity. She noted that future studies will explore a 1 x 107 dose level, intensification of lymphodepletion prior to CAR-T infusion, and consideration of the potential role of immune checkpoint inhibitors to augment CAR-T in extramedullary disease.

Read more of Cancer Therapy Advisors coverage of AACR 2020 meeting by visiting the conference page.

References

Shalabi H, Yates B, Shahani S, et al. Safety and efficacy of CD19/CD22 CAR T cells in children and young adults with relapsed/refractory ALL. Presented at: American Association for Cancer Research (AACR) Virtual Annual Meeting I 2020; April 27-28, 2020. Abstract CT051.

Originally posted here:
Novel Bispecific CD19/CD22 CAR-T Therapy Deemed Tolerable in Relapsed/Refractory ALL - Cancer Therapy Advisor

Targeted pathological collagen delivery of sustained-release rapamycin to prevent heterotopic ossification – Science Advances

Abstract

Heterotopic ossification (HO) in connective tissues like tendons and ligaments severely damages tissue structure. The pathogenesis of HO remains unclear but may involve mTOR. The results presented here indicate that tendon stem/progenitor cells do not undergo osteochondrogenic differentiation when mTOR signaling is inactivated by gene knockout or rapamycin (RAPA) treatment. Meanwhile, it is necessary to deliver RAPA to the injured sites and avoid disturbing the normal tendon. A RAPA delivery system, developed using collagen hybrid peptide (CHP) to modify the surface of poly(lactic-co-glycolic acid) (PLGA) nanoparticles, targeted RAPA specifically to pathological tendon collagen. The CHP-PLGA-RAPA nanoparticles showed excellent pathological collagen affinity, sustained-release ability, and bioactivity. In a mouse model of tendon HO, CHP-PLGA-RAPA nanoparticles specifically bound to pathological tendon and strongly suppressed HO progression. The mTOR signaling pathway appears to be a viable therapeutic target for tendon HO, and CHP-PLGA nanoparticles may be valuable for the treatment of tendon-related diseases.

Approximately 30% of clinical consultations for musculoskeletal system diseases involve tendon injury (1). The tendon lacks cells and blood vessels, so its ability for endogenous repair is inadequate. For this reason, most cases of tendon injury progress to tendinopathy. The clinical symptoms of chronic tendinopathy mainly manifest as pain, swelling, and inability to perform normal activities (24). One subtype of tendinopathy is tendon heterotopic ossification (HO), which damages the structure of the tendon tissue itself, aggravates symptoms, and severely impairs tendon function (5).

Endochondral ossification can include the development of ectopic calcification in connective tissues, including tendons (6, 7). A previous study has demonstrated that injured tendons in humans express higher levels of cartilage-associated matrix proteins and marker genes (8). In addition, tendon stem/progenitor cells (TSPCs), when localized in the inflammatory niche of tendinopathy, exhibit a strong tendency to differentiate into chondroid and osteoid tissues (9). Consequently, we hypothesized that early therapeutic intervention in tendon injury could prevent the onset of inappropriate TSPC differentiation, thereby protecting tendons from ectopic ossification. Currently, the main challenges are the lack of knowledge regarding the critical ossification mechanisms and the absence of an appropriate system to deliver drugs precisely to the pathological tendon.

Ectopic osteogenesis in a variety of tissues may be under the control of the mammalian target of rapamycin (RAPA) (mTOR) signaling pathway, a crucial regulator of cell biological processes, including cell growth, metabolism, and protein synthesis (1013). Several studies have implicated mTOR in tendon and ligament ossification, although different perspectives have prevented a consensus on underlying mechanisms. For example, Jiang et al. (14) reported that mTOR inhibition can decelerate rat tendon ossification after Achilles tenotomy. By contrast, another transcriptome-level investigation demonstrated down-regulation of mRNA expression of the mTOR signaling pathway in human ossified ligament (15), but up-regulation of circular RNA expression (15). For these reasons, further exploration of mTOR function in tendon HO is essential.

In the present study, tendon tissuespecific mTOR gene knockout mice were used to demonstrate the essential function of mTOR signaling in the progression of tendon HO. We hypothesized that the inhibitor RAPA could prevent tendon HO by modulating the abnormal differentiation of TSPCs. However, we recently found that mTOR also mediates the normal tenogenic differentiation of TSPCs (16). Accordingly, direct injection of RAPA might be unsuitable for treating tendinopathy because tendon injury or tendon ossification usually occurs in a local position and not along the whole tendon.

We have fabricated a drug delivery system that can target and bind to injured collagen. When a lesion appears on a tendon, inflammatory factors initiate the expression of matrix metalloproteinase, with a resulting degradation of the extracellular matrix and damage to the inherent triple helix structure of collagen (1, 17). Collagen hybrid peptide (CHP) is a synthetic molecule that specifically binds to this intact and denatured collagen structure (1820). We devised a drug delivery system consisting of nanoparticles made from poly(lactic-co-glycolic acid) (PLGA), a biocompatible and biodegradable material, with surfaces covered with CHP. This system facilitated the accurate positioning and integration of PLGA onto a pathological tendon while avoiding healthy tendons. CHP-PLGA nanoparticles containing encapsulated RAPA were able to provide a sustained release of RAPA to treat the pathological tendon of a mouse model of HO.

The findings presented here provided new insights into the mechanism of tendon HO and established a powerful drug delivery system for treatment of tendon-related diseases. The targeted delivery of RAPA to pathological collagen could be a therapeutic approach for prevention of HO. It may also conceivably be used as an effective approach to prevent and treat other diseases associated with the destruction of collagen structure.

A mouse Achilles tendon HO model was induced by collagenase in this study. The inflammation followed the breakdown of the collagen matrix in the early stage and then receded because of the rodents robust self-healing ability. Immunohistochemical staining indicated a marked increase in the expression of interleukin-1 (IL-1) at 1 week after tendon injury, followed by a return to a near-normal level at 3 weeks (fig. S1A). However, the emergence of inflammation inevitably induced incorrect TSPC differentiation to eventually trigger HO formation. We then established Scx-cre;mTmG mice whose cells expressed red fluorescent protein (tdTomato) before exposure to Cre recombinase, while Cre-expressing cells and their derivatives would stop to express green fluorescent protein (GFP). Immunofluorescence (IF) following induction of the HO model with collagenase in these Scx-cre;mTmG mice revealed colocalization of GFP and the osteogenic differentiationrelated gene bone morphogenic protein 2 (BMP2) (fig. S2A). GFP also colocalized with the chondrogenic differentiationrelated gene SOX9 (fig. S2B). These findings indicated that TSPCs participated in the formation of HO under inflammatory conditions.

The role of mTOR in tendon HO was investigated further in a transgenic mouse model in which the mTOR gene was conditionally knocked out in the tendon lineage (for convenience, the Scx-Cre;mTORfloxed mice are henceforth referred to as mTOR-TKO, for mTOR tendon knockout mice) (16). Collagenase was then injected into the Achilles tendon of mTORfloxed [termed wild-type (WT) here for simplicity] mice or mTOR-TKO mice to obtain the HO model in both mouse types. After 6 weeks, significant high-intensity zones were observed on microcomputed tomography (micro-CT) images of Achilles tendon in the WT group (Fig. 1A). By contrast, the mTOR-TKO mice showed virtually none of these zones (Fig. 1B). Histological staining results, including hematoxylin and eosin (H&E), Masson trichrome (Masson), and Safranin O (SO) staining, also showed typical endochondral ossification in the WT mouse tendons, but not in the mTOR-TKO mouse tendons (Fig. 1, C to F). Isolation of TSPCs from WT and mTOR-TKO mice and culture in an osteogenic induction medium revealed reduced alkaline phosphatase (ALP) and Alizarin Red S (ARS) staining in the TSPCs from mTOR-TKO mice, indicating a weaker osteogenic differentiation ability than in the WT mice (Fig. 1, G to J). The TSPCs from mTOR-TKO mice also grew more poorly than WT TSPCs in the chondrogenic induction culture system (Fig. 1K). These findings confirmed that the osteogenesis and chondrogenesis ability of the injured tendon was sharply decreased at the tissue and cell level by knockout of mTOR signaling.

(A and B) Micro-CT imaging and quantitative analysis of the Achilles tendon from WT and mTOR knockout (mTOR-TKO) mice. n = 10. Scale bar, 2 mm. (C to F) HE, Masson, and SO staining and histological score evaluation of WT and mTOR-TKO mice. n = 10. Scale bar, 50 m. (G and H) ALP staining and positive rate of cells cultured in an osteogenic medium for 7 days. n = 3. Scale bars, 100 m. (I and J) ARS staining and optical density (OD) value of cells cultured in the osteogenic medium for 14 days. n = 3. Scale bar, 200 m. (K) Light microscopy of cells cultured in the chondrogenic medium for 3 days. BV, bone volume.

We then investigated potential molecular mechanisms involved in ossification via the mTOR signaling pathway by conducting RNA-sequencing (RNA-seq) studies on the tendon tissues. We used the WT mice as a reference and found 1261 down-regulated genes and 611 up-regulated genes in the mTOR-TKO group (fold change, 2; q value < 0.05) (Fig. 2A), which was consistent with the current knowledge about the function of the mTOR signaling pathway and its primary relationship with cell growth and anabolism (10). A subsequent comparison of the gene expression differences between the WT and mTOR-TKO mice (Fig. 2B and fig. S3A) and gene ontology (GO) analysis revealed that mTOR tendon knockout weakened cell osteogenesis and chondrogenesis, and it down-regulated genes related to tissue mineralization and ossification (Fig. 2C). Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that the top 20 down-regulated items in mTOR-TKO mice were those related to PI3K (phosphatidylinositol 3-kinase)AKT, mitogen-activated protein kinase, insulin, and other signaling pathways (fig. S3B) reportedly involved in ectopic ossification development in tendons (2124). A gene set enrichment analysis (GSEA) was also performed on all genes because of the inherent limitations of differentially expressed gene analysis. The results demonstrated that mTOR signaling was indeed down-regulated in the mTOR knockout mice and that the BMP signaling pathway and the response to BMP signals were both notable down-regulated (Fig. 2, D and E, and fig. S4A). BMP signaling is recognized as a crucial regulator of osteogenic differentiation of TSPCs (25, 26). Overall, these in vivo and in vitro findings supported a role for mTOR signaling pathway in tendon HO.

(A) Volcano plot of gene expression (mTOR-TKO versus WT; fold change, 2; q value < 0.05). (B) Heat map of differentiated expression genes. (C) GO analysis of differentially expressed genes. (D and E) GSEA of the genes associated with mTOR signaling and BMP signaling. NES, normalized enrichment score; FDR, false discovery rate; KO, knockout.

An in vitro cell assay confirmed that 2 nM RAPA had a negligible effect on cell proliferation but effectively inhibited the expression of p-mTOR and p-S6 (Fig. 3, A and B). Stimulation of TSPCs in chondrogenic culture with IL-1 resulted in intensification of both Alcian blue and SO staining when compared with an unstimulated control group, suggesting an augmented expression of cell materials containing anionic groups, such as chondroitin sulfate and keratan sulfate (Fig. 3, C, D, G, and H). In other words, the ability of TSPCs to differentiate into chondrocytes was enhanced. The addition of RAPA blocked the effect induced by IL-1 (Fig. 3, C, D, G, and H). Similarly, when we used osteoinductive medium to induce osteogenesis of TSPCs, RAPA suppressed osteogenic differentiation of TSPCs induced by the IL-1 stimulation, as revealed by ALP and ARS staining (Fig. 3, E, F, I, and J). Human TSPCs showed similar RAPA responses (fig. S5, A to J). RAPA therefore prevented the abnormal differentiation of TSPCs under inflammatory stimulation by inhibiting the mTOR signaling pathway.

(A) Cell proliferation of TSPCs when cultured with different concentrations of RAPA. (B) The protein expression of mTOR signaling pathway, including p-mTOR, mTOR, p-S6, S6, and the reference protein -tubulin. (C and D) Alcian blue and SO staining of TSPCs cultured in the chondrogenic medium for 14 days. Scale bars, 50 m. (E and F) ALP and ARS staining of TSPCs cultured in the osteogenic medium for 7 and 14 days, respectively. Scale bars, 50 and 200 m, respectively. (G) Optical density (blue) of Alcian blue staining. n = 3. ns, not significant. (H) Optical density (red) of SO staining. n = 3. (I) Positive rate of ALP staining. n = 3. (J) Optical density value of ARS staining. n = 3.

The mTOR pathway is critical for tendon development, as confirmed by the tendon defects observed due to tendon-specific ablation of mTOR in our mTOR-TKO mice and the impaired tenogenesis of mesenchymal stem cells by RAPA (16). The RNA-seq analysis results also indicated that the transforming growth factor (TGF-) signaling pathway was down-regulated in mTOR-TKO mice (fig. S4B). Similar changes were also observed in tendon-related genes (fig. S4C). TGF- controls a bidirectional regulatory pathway that participates in the process of tendon HO and in the promotion of tenogenesis in mesenchymal stem cells (27, 28). We hypothesized that these dual actions of mTOR could also regulate the behavior of TSPCs. Treatment of TSPCs with high concentrations of RAPA inhibited TSPC proliferation, but the inhibitory effect disappeared upon withdrawal of RAPA (fig. S6A). RAPA at 2 nM also inhibited the expression of tendon-related genes in the TSPCs, and this expression was rapidly restored after drug withdrawal (fig. S6B). These findings indicated that the effects of RAPA were reversible.

CHP-PLGA nanoparticles were synthesized to provide targeted delivery and sustained release of RAPA at sites of tendon damage. The PLGA nanoparticles were prepared using an oil/water (O/W) emulsion-solvent evaporation method with bovine serum albumin (BSA) molecules acting as stabilizers in the water phase. The BSA molecules covering the particle surface enabled the subsequent covalent conjugation of a peptide with amino groups using glutaraldehyde as a cross-linker. The obtained PLGA nanoparticles were spherically shaped, with diameters ranging from 100 to 300 nm in the dry state, as observed in scanning electron microscopy (SEM) and transmission electron microscopy (TEM) images (Fig. 4, A and B). The surface zeta potential of the PLGA nanoparticles was 18.9 mV (Fig. 4C) due to the surface cover with negatively charged BSA. The hydrophilic diameter of the PLGA nanoparticles was 227 38 nm (Fig. 4D), which was larger than the dry-state diameter because of the hydration of the BSA surface coating.

(A and B) TEM and SEM images of PLGA and CHP-PGLA nanoparticles. Scale bars, 1 m. (C) Zeta potential of PLGA and CHP-PLGA nanoparticles. n = 4. (D) Size distribution of PLGA and CHP-PLGA nanoparticles. (E and F) CHP density at various feeding concentrations and different times. n = 4. (G) Drug release from PLGA and CHP-PLGA nanoparticles over the course of 10 days. n = 4. (H) TEM of CHP-PLGA binding to intact and injured collagen. (I) SEM of CHP-PLGA binding to intact and injured tendon.

As shown in Fig. 4 (E and F), the densities of immobilized CHP peptide molecules on the CHP-PLGA nanoparticles increased with increasing concentrations of the reagent in the feeding solution and with increasing reaction time. Specifically, the density of CHP reached 4.2 104 molecules per nanoparticle (i.e., approximately 0.6 molecule/nm2) at a peptide feeding concentration of 80 g/ml and a reaction time of 16 hours. These conditions produced a sufficiently high CHP density, so they were used in subsequent experiments.

The introduction of CHP onto the surface did not produce any obvious changes in the morphology, size, or surface zeta potential of the PLGA nanoparticles (Fig. 4, A to D). The loading ratios of RAPA for both PLGA and CHP-PLGA nanoparticles were quite similar (39 and 37 g/mg, respectively) when subjected to the same incubation and washing processes. Approximately 14.7% of the loaded RAPA was released during the peptide conjugation process because this step takes a long time at room temperature. This release is actually advantageous since it enables a sustained release of RAPA because the initial burst release has already occurred. Both PLGA and CHP-PLGA showed a good sustained release of encapsulated RAPA, as determined by almost linear release curves versus time for more than 1 week (Fig. 4G). The RAPA was gradually released because of the degradation of the polymer matrix and its slow diffusion into the medium.

The ability of CHP-PLGA to target-bind to injured collagen was then evaluated in vitro by two methods (i.e., incubation of CHP-PLGA with collagen denatured by high temperature or with tendon damaged with collagenase). SEM and TEM observations revealed that the CHP coating aided the specific attachment of the CHP-PLGA nanoparticles to injured collagen or injured tendon, as attachment was rarely observed to intact collagen or intact tendon (Fig. 4, H and I). Incubation of PLGA and CHP-PLGA nanoparticles with normal and diseased human tendon also revealed binding of a large amount of CHP-PLGA to pathological collagen, but only a very small amount of nonspecific binding to healthy tendon, indicating a therapeutic potential of these nanoparticles in clinical practice (fig. S7A).

We then examined the biological activity of CHP-PLGA-RAPA nanoparticles. The culture medium was changed every other day, and the amount of RAPA released from the CHP-PLGA-RAPA nanoparticles was approximately 25% on the second day. Therefore, we increased the concentration of CHP-PLGA-RAPA to 8 nM to ensure that the total amount of RAPA released the next day was approximately equal to the amount previously used. The RAPA released from CHP-PLGA maintained its biological effect (i.e., it inhibited the mTOR signaling pathway, osteogenesis, and chondrogenesis of TSPCs following inflammatory stimulation; fig. S8, A to J). The CHP-PLGA-RAPA nanoparticles therefore showed potential as a drug delivery system for tendon disease with sustained-release ability, targeted binding, and biological function.

The drug delivery efficiency of CHP-PLGA nanoparticles in pathological mouse tendon was evaluated by synthesizing CHP-PLGA-Cy5 nanoparticles and injecting them into mouse tendons after HO modeling. The replacement of RAPA with Cy5 allowed the detection of the nanoparticle cargo with infrared fluorescence imaging devices to provide general feedback regarding drug retention. The CHP-PLGA nanoparticles significantly enhanced the retention of Cy5 in the tendon tissue at various time points when compared with PLGA nanoparticles, indicating that the CHP coating provided a stronger affinity for damaged tendon and made the nanoparticles a more effective drug delivery vehicle (Fig. 5, A to C).

(A) In vivo fluorescence images of injured Achilles tendon after subcutaneous injection of nanoparticles. Scale bars, 1 cm. (B and C) Total radiant efficiency and percentage of signal preservation of nanoparticles at different time points. n = 5.

The HO model causes notable inflammation, so drug administration was halted at 3 weeks to allow the tendon tissue to pass through the inflammatory window without hindering the subsequent tendon repair process (Fig. 6A). The immunohistochemical results suggested a substantial down-regulation of the mTOR signaling pathway in the PLGA-RAPA and CHP-PLGA-RAPA treatment groups at 3 weeks (fig. S9A). At 6 weeks, micro-CT imaging indicated that PLGA-RAPA and especially CHP-PLGA-RAPA significantly decreased the occurrence of HO in the mouse tendons (Fig. 6, B and C). All histological evaluations, including HE, Masson, and SO staining, identified significant improvement in the sustained-release medication group. Moreover, little or no ectopic bone tissue was observed following CHP-PLGA-RAPA or PLGA-RAPA treatment (Fig. 6, D to G). The primary difference was that CHP-PLGA-RAPA almost completely inhibited endochondral ossification, whereas some chondroid tissues, which were not apparent in micro-CT images, appeared in the PLGA-RAPA group. Overall, the CHP-PLGA-RAPA nanoparticles showed good tissue-specific binding ability and were effective in preventing abnormal differentiation of TSPCs caused by an inflammatory microenvironment.

(A) Animal experiment design. (B and C) micro-CT images of the Achilles tendon at 6 weeks after HO modeling and quantitative analysis. n = 10. Scale bar, 2 mm. (D to G) HE, Masson, and SO staining and histological score evaluation of the Achilles tendon at 6 weeks. n = 10. Scale bars, 50 m.

In this study, mTOR signaling was found to play a dominant role in tendon ectopic osteogenesis occurring both in vitro and in vivo. Because mTOR inhibition also influences the physiological tenogenesis of normal tendon, we established a mouse model of tendon HO, and we developed a drug delivery system that would target the diseased tendon tissue to reduce side effects on normal tendon. This drug delivery system almost completely inhibited the development of tendon HO in the mouse model.

Until now, our understanding of the mechanism underlying tendon HO has been limited. However, some studies have highlighted a possible involvement of mTOR in the process of tendon HO. For example, Hino et al. (12, 29) reported that mTOR signaling up-regulated aberrant chondrogenesis in fibrodysplasia ossificans progressiva disease. Similarly, Xu et al. (13) identified the AKT-mTOR pathway as a therapeutic target for the treatment of inflammation-mediated osteogenesis of stem cells in periodontal ligament. A rat HO model induced by Achilles tendon transection also indicated that RAPA modulated the osteogenesis of TSPCs (14). In the present study, the combination of an in vitro cell experiment with RNA-seq analysis and conditional gene knockout mice revealed that mTOR was an obligatory link in the tendon endochondral ossification process. We observed that cells expressing the Scx gene were involved in ectopic osteogenesis, in agreement with previous research (25, 26). The Scx gene serves as a marker for TSPCs, thereby confirming the participation of endogenous stem cells from the tendon in the development of tendon HO.

The levels of IL-1 notable increased in the disease model, indicating that inflammation initiated the abnormal differentiation of TSPCs. We also observed a temporary increase in the expression of IL-1 in tendon injury, which led to the final tendon ossification. These findings suggested that once stem cells are exposed to external stimuli, the activated differentiation process is irreversible. Hence, in patients with HO, more attention must be paid to preserving the stemness of endogenous stem cells while taking measures to block extrinsic causes of abnormal differentiation.

In this study, we exploited a tendinopathy characteristic, namely, that collagen would degrade following an injury, and we used CHP-PLGA as a drug carrier to deliver targeted slow-release RAPA to an injured tendon. To the best of our knowledge, this is the first documented use of CHP as a targeting tool with potential for treatment of tendon disease. When compared with traditional direct drug injection, this delivery system performs both sustained-release and diseased tissue target-binding functions. The sustained-release property would greatly reduce the required frequency of drug injections in clinical practice. The specific binding of PLGA to pathological collagen imparted by the CHP coating means that patients could be given subcutaneous injections, thereby avoiding secondary injury due to tendon puncture. Microscopy observations indicated that CHP-PLGA may be more concentrated in and near the lesion, rather than randomly distributed throughout the tendon. If this also proves to be the case in a clinical situation, then this would substantially improve the sensitivity and effectiveness of the treatment and decrease possible side effects on normal tendons.

We speculated that the interaction of CHP with pathological tendon tissue was why CHP-PLGA exhibited greater potency than PLGA in the treatment of ectopic ossification of tendons, even though the PLGA-Cy5 study confirmed that PLGA nanoparticles were equally effective at retaining drugs. Moreover, unlike other antibodies that can only specifically target a certain type of collagen, CHP can target any type of denatured collagen chains and triple helix structures (30). In our study, HO was induced by collagenase, confirming that the collagen damage targeting mechanism works effectively.

Trauma in connective tissues is known to induce HO formation (31), and CHP would also be expected to target tendon damage due to trauma because injury would trigger an inflammatory reaction. Inflammatory factors, such as IL-1 and tumor necrosis factor, activate matrix metalloproteinases that degrade collagen, thereby providing CHP with a target. Fibrodysplasia ossificans progressiva is a known HO disease of muscle and connective tissues (29), but HO in that disease is attributed to a mutation in the ACVR1 gene, with a pathological process unrelated to collagen fiber damage. In that type of collagen disease, the use of CHP may not be an effective strategy. Nevertheless, derivatives of CHP may also be available for the diagnosis and treatment of collagen degenerationrelated diseases, such as osteoarthritis and intervertebral disc disease.

The Achilles tendon in C57BL/6 mice and the remaining tendon graft tissues from autologous human tendon transplantation (i.e., from anterior cruciate ligament reconstruction surgery) were cut into pieces, digested with 0.2% type 1 collagenase, screened by low-density culture, and lastly cultured in a basic medium [low-glucose Dulbeccos modified Eagles medium (L-DMEM), 10% fetal bovine serum (FBS), and 1% penicillin-streptomycin].

The osteogenic medium consisted of high-glucose DMEM (H-DMEM) containing 108 M dexamethasone, vitamin C (50 g/ml), 10 mM -glycerol phosphate, and 10% FBS. On day 7 of culture, the ALP activity of TSPCs was tested with a commercial detection kit (Beyotime, C3206). Cell nuclei were then stained with 4,6-diamidino-2-phenylindole to count the total cell numbers and calculate the positive rates of ALP staining. On day 14, the calcium deposits formed by TSPCs were stained with ARS (Sigma-Aldrich, A5533). The stained nodules were solubilized with 5% SDS in 0.5 N HCl for 30 min at room temperature, and the optical density of the solution was determined at 405 nm.

The chondrogenic medium consisted of H-DMEM containing 1 mM sodium pyruvate, 1% Insulin-Transferrin-Selenium, vitamin C (50 g/ml), 107 M dexamethasone, and TGF-3 (10 ng/ml). Chondrogenesis was evaluated in a micromass culture system described previously (32). At day 14, cells were stained with Alcian blue or SO. The optical density (blue for Alcian blue and red for SO) of the microphotograph was analyzed using image analysis software (IPP 6.0).

The tenogenic medium was H-DMEM containing vitamin C (50 g/ml) and 10% FBS. After the cells reached nearly 90% confluence, the basic medium was replaced with the induction medium, culture was continued for 14 days, and the TSPCs were then collected for RNA extraction and quantitative real-time polymerase chain reaction (qRT-PCR) analysis.

Cell proliferation was measured with a commercial kit (CCK-8, Dojindo, CK04). Briefly, TSCPs were incubated in a 10% CCK-8 solution at 37C for 1 hour. The optical density of the solution was then measured at 450 nm with a microplate reader (SpectraMax 190, Molecular Devices).

TSPCs were lysed with TRIzol (Takara, 9109) to harvest total RNA, which was reverse-transcribed into complementary DNA (cDNA). The qRT-PCR was performed with a SYBR Green Mix (Takara, RR420A) in a fluorescence signal detection device (Roche, LightCycler 480).

The primers used in this analysis were the following:

The antibodies used in this study were the following: rabbit anti-mTOR [Cell Signaling Technology (CST), 2983], rabbit antip-mTOR (CST, 5536), rabbit anti-S6 (CST, 2217), and rabbit anti-pS6 (CST, 4858).

An ultrasonic liquid processor (MISONIX, USA) was used throughout the study to prepare PLGA (poly(lactic-co-glycolic acid) (lactic acid:glycolic acid = 75:25, Mw, ~78 kDa, Jinan Daigang Biomaterial Co. Ltd., China) nanoparticles using BSA (Sigma-Aldrich) as the emulsifier. Briefly, PLGA and RAPA (S1039, Selleck) were dissolved in dichloromethane at 20 and 1 mg/ml, respectively. A 1-ml volume of the PLGA/RAPA mixture was quickly added to 4 ml of emulsifier solution (3% BSA, aqueous solution) under vigorous magnetic stirring, followed by probe sonication at 10 W for 30 s on ice to complete the emulsification. This O/W emulsion was then dispersed into a 20-fold volume of Milli-Q water and stirred for at least 3 hours until it solidified. The resulting BSA/PLGA nanoparticles were then collected, purified by centrifugation at 12,000 rpm for 10 min, and resuspended in Milli-Q water three times to remove residual nonincorporated BSA molecules.

CHP was covalently conjugated to the BSA-coated PLGA nanoparticles using glutaraldehyde chemistry. Before preparation, the helix structure of the CHP peptide was disassembled by heating to 80C for 5 min and then cooling on ice for 1 min. The suspension of PLGA nanoparticles (1 mg/ml) was mixed with glutaraldehyde (final concentration of 1 mg/ml) for 12 hours at room temperature under gentle shaking. During this step, one aldehyde group from the glutaraldehyde molecule reacted with amino groups from the BSA on the surface of PLGA nanoparticles. Since the amount of glutaraldehyde was in excess of that of BSA [PLGA nanoparticles (about 50 g/mg)], the cross-linking of BSA molecules was avoided. After centrifugation and washing again with water, the CHP peptide was added to the system. Further conjugation was allowed to occur between amino groups from peptide and the aldehyde groups on the PLGA nanoparticles for a certain period of time at room temperature. The peptide-modified PLGA nanoparticles were collected by centrifugation at 12,000 rpm for 10 min and washed with water three times to remove unbound CHP peptide molecules.

The fluorescence intensity of a fluorescein isothiocyanate (FITC)labeled peptide solution before and after this reaction was compared to allow fluorescence spectroscopy assessment (LS55, PerkinElmer, USA) of the amount of bound peptide. In a typical experiment, PLGA nanoparticles (1 mg/ml) were activated with glutaraldehyde and then mixed with FITC-labeled peptide for conjugation. The initial solution volume and fluorescence intensity of the solution were recorded and denoted as Vi and Fi, respectively. After reaction and washing, all the medium was collected. This second volume and corresponding fluorescence intensity were designated as Va and Fa, respectively. The amount of conjugated peptide was calculated according to the equation: m = ViFi VaFa, and was determined by reference to a standard curve derived from fluorescence intensity versus peptide concentration.

The concentration of the different PLGA nanoparticles was determined by weighing the dried particles obtained from 1 ml of the suspension solution. The PLGA nanoparticle solution (100 g/ml) was then used to measure the hydrodynamic diameters and the surface zeta potential in Milli-Q water (Beckman Delsa Nano, Beckman Coulter). The morphology of PLGA nanoparticles was observed by SEM and TEM.

The release of RAPA from the PLGA nanoparticles was monitored in phosphate-buffered saline (PBS; pH 7.4) at 37C under gentle shaking. In brief, 1 ml of PLGA nanoparticles (1 mg/ml) was transferred to a dialysis bag (10-kDa cutoff; Sigma-Aldrich) and immersed into 10 ml of release buffer. At predetermined time intervals, 0.5 ml of release buffer was removed for measurement, and the same amount of fresh buffer was added back. The concentration of released RAPA was determined with a high-performance liquid chromatography system equipped with a Diamonsil C18 column (150 mm by 4.6 mm, 5 m; Dikma Technologies Inc., Lake Forest, CA, USA) and a Hitachi L-2130 pump and L-2400 ultraviolet (UV) detector (Hitachi, Tokyo, Japan). The mobile phase was methanol and water (75:25, v/v) delivered at a flow rate of 1.0 ml/min. The UV detection wavelength was 278 nm, the column temperature was 50C, and the injection volume was 20 l.

Type 1 collagen (0.5 mg/ml in PBS) was denatured by heating at 80C for 5 min and then placed in an ice bath. Mouse tendons were damaged by digestion with 2% collagenase for 30 min in a 37C incubator. A 10-l sample of injured type 1 collagen or 10 l of intact type 1 collagen was added to a TEM grid. After 1 min, the excess solution was removed with blotting paper, and 10 l of PLGA or CHP-PLGA nanoparticles (10 nM) was applied. The grid was incubated for 10 min and then washed three times with double-distilled water. The final sample was stained with 2% (w/v) uranyl acetate by applying 10 l of staining solution to the grid and removing the excess solution with blotting paper after 1 min. The grid was dried at room temperature overnight and then observed by TEM. Intact tendons and injured tendons were subsequently incubated with CHP-PLGA (10 M) for 2 hours on a shaking table at room temperature. The tendons were washed with PBS three times and further processed for SEM.

Animal experiments were conducted with the approval of the Zhejiang University Experimental Animal Welfare and Ethics Committee under Institutional Animal Care and Use Committee guidelines (ZJU20190049). The tendon HO model was established by injecting type 1 collagenase (12.5 U per leg; Gibco, 17100017) into the midpoint of the right Achilles tendons of 8-week-old mice, and the same operation was repeated 3 days later to stabilize the effect. Ten WT mice and 10 mTOR-TKO mice were euthanized at 6 weeks after modeling, and the legs were removed for micro-CT imaging and subsequent histology.

In an in vivo drug experiment, 60 mice were divided equally into four groups, and each mouse received a subcutaneous injection (between Achilles tendon and skin) once per week: control group (sterile PBS, 5 l), RAPA group (RAPA, 70 M, 5 l), PLGA-RAPA group (70 M, 5 l), and CHP-PLGA-RAPA group (70 M, 5 l). At 3 weeks after treatment, five mice in each group were euthanized for immunohistochemistry (IHC). At 6 weeks after treatment, the remaining 10 mice in each group were euthanized, and the legs were taken for micro-CT imaging and subsequent histology.

In an in vivo cell tracing experiment, three WT mice and three Scx-cre;mTmG mice were euthanized at 6 weeks after modeling. Achilles tendons were then isolated and prepared in paraffin sections for IF studies.

Hindlimbs from the mice were fixed in 4% paraformaldehyde and analyzed by micro-CT (Skyscan 1172). The scanner was set at a voltage of 80 kV and a resolution of 18 m per pixel. The images were reconstructed, analyzed for HO bone volume, and visualized by NRecon, CTAn, and CTVol.

RAPA was replaced with Cy5 (Lumiprobe, 23020) in the synthesis process of PLGA-RAPA or CHP-PLGA-RAPA to obtain PLGA-Cy5 or CHP-PLGA-Cy5 nanoparticles, respectively. At 1 week after HO modeling, mice were injected with PLGA-Cy5 or CHP-PLGA-Cy5 and then anesthetized with isoflurane. The in vivo nanoparticle distribution was analyzed with a fluorescence imaging system (IVIS SpectrumCT, PerkinElmer) at the indicated time points.

Tissue specimens were fixed in 4% paraformaldehyde, washed with running water, dehydrated in a graded ethanol series, vitrified with dimethylbenzene, and embedded in paraffin. Paraffin sections (7 m) were deparaffinized in xylene, hydrated with gradient ethanol, and stained with standard H&E, SO, or Masson staining procedures. Histological scores were calculated from the results of H&E staining.

For IHC or IF analysis, sections were incubated at 4C overnight with primary antibodies. The following primary antibodies were used in this study: rabbit anti-GFP (CST, 2555), mouse anti-GFP (CST, AG281), rabbit anti-BMP2 (Beyotime, AF0075), mouse anti-SOX9 (Abcam, ab76997), rabbit anti-mTOR (CST, 2983), rabbit antip-mTOR (CST, 5536), rabbit anti-S6 (CST, 2217), rabbit anti-pS6 (CST, 4858), and rabbit antiIL-1 (Abcam, ab9722). For IHC, the sections were also incubated with horseradish peroxidaselinked secondary antibodies (CST, 7074) for 1.5 hours, and the staining was visualized with 3,3-diaminobenzidine solution (ZSGB-BIO, ZLI-9017). For IF, sections were subsequently incubated with fluorescein-conjugated secondary antibodies for 1.5 hours and observed under a confocal fluorescence microscope (Nikon A1R, Japan).

Tendon tissues from WT and mTOR-TKO mice were ground in TRIzol to prevent RNA degradation. After verification of the RNA integrity, the extracted RNA was reverse-transcribed to create a cDNA library for subsequent sequencing.

The quantitative data were presented as means SD. Students t test was performed to assess whether statistical differences existed between groups. Multiple comparisons were performed with a one-way analysis of variance (ANOVA) and Tukeys post-test. P values <0.05 were considered statistically significant. The significance level is presented as *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001.

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

Acknowledgments: Funding: This work was supported by the National Key Research and Development program of China (2018YFC1105100 and 2016YFE0132700), the NSFC grants (81972099, 81522029, 81772418, 31570987, 81871764, 81572157, 51822306, 31571402, 81572115, and 81874019), the Zhejiang Provincial Natural Science Foundation of China (LR20H060002), and the Fundamental Research Funds for the Central Universities (2019QNA7040). We are grateful to the Core Facilities of Zhejiang University School of Medicine, and Bio-Ultrastructure Analysis Lab. of Analysis Center of Agrobiology and Environmental Sciences of the Zhejiang University for technical assistance. Author contributions: Conception: Y.C., W.S., Y.Z., Z.M., and X.C. Research design: Y.C., W.S., and X.C. Data acquisition/analysis: Y.C., W.S., C.T., J.H., C.F., Z.Y., Y.H., and Z.M. Resource assistance: W.C., H.O., Y.Z., Z.M., and X.C. Writing, drafting, and editing: Y.C., W.S., Y.Z., Z.M., and X.C. Competing interests: The authors declare that they have no 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.

More:
Targeted pathological collagen delivery of sustained-release rapamycin to prevent heterotopic ossification - Science Advances

First-in-Human Universal CAR-T Therapy Found Active in Relapsed/Refractory T-ALL – Cancer Therapy Advisor

Thefirst-in-human, universal chimeric receptor antigen (CAR) T-cell (CAR-T)therapy GC027 was tolerable and resulted in antileukemic responses amongpatients with relapsed/refractory T-cell acute lymphoblastic leukemia (T-ALL),according to results from a phase 1 trial presented at the American Associationfor Cancer Research (AACR) Virtual Annual Meeting I 2020.1

Theuniversal CAR T cells target CD7, which, according to Xinxin Wang, PhD, ofGracell Biotechnologies Co, Ltd, in China, and lead author and presenter of thestudy, is a good target for T-ALL because it is expressed by more than 95% ofT-ALL patients.

GC027 isallogeneic, which may prevent the development of graft-versus-host disease. Theproduct is introduced using lentivirus for rapid elimination of T-ALL cells. Preclinicalstudies showed efficacy in a T-ALL xenograft model, and this prospective studyevaluated the safety and efficacy in humans.

Thesingle-arm, open-label study treated 5 adult patients with relapsed/refractoryCD7-positive T-ALL with a single infusion of 1 of 3 different dose levels ofG027: 0.6 x 107/kg, 3 x 107/kg, and 1.5 x 107/kg.Lymphodepletion therapy was administered prior to the G027 infusion. Theprimary endpoint was safety and the secondary endpoints included objectiveresponse rate (ORR) within 3 months after G027 infusion.

Patientswith extramedullary or central nervous system disease were excluded. Atbaseline, the median age was 24 (range, 19-38). Patients were heavilypretreated, with 5 median number of prior therapies (range, 1-9). Two patientshad high-risk disease and the median bone marrow tumor burden was a median of38.2% of blasts. None of the patients had undergone a prior allogeneic hematopoieticstem cell transplant.

Allpatients developed cytokine release syndrome (CRS), 4 of which were grade 3 and1 was grade 4. All cases were manageable and resolved with treatment andsupportive care. None of the patients developed neurotoxicity.

The completeremission (CR)/CR with incomplete hematologic recovery was 100%. By day 28, 4patients achieved a CR with negative for minimal residual disease (MRD) and 3of these patients remained MRD negative up to day 161. One patient achieved CRbut was MRD positive, and relapsed by day 29.

Peak CART-cell expansion in peripheral blood occurred between week 1 and 2.

As the first-in-human, universal CAR T-cell therapy for adult relapsed/refractory T-ALL, Dr Wang said, GC027 has demonstrated superior clinical efficacy and induced deep response in patients with acceptable safety profile. She added that trial enrollment is ongoing.

Read more of Cancer Therapy Advisors coverage of AACR 2020 meeting by visiting the conference page.

Reference

Wang X, Li S, Gao L, et al. Clinical safety and efficacy study of TruUCAR GC027: The first-in-human, universal CAR-T therapy for adult relapsed/refractory T-cell acute lymphoblastic leukemia (r/r T-ALL). Presented at: American Association for Cancer Research (AACR) Virtual Annual Meeting I; April 27-28, 2020. Abstract CT052.

Excerpt from:
First-in-Human Universal CAR-T Therapy Found Active in Relapsed/Refractory T-ALL - Cancer Therapy Advisor

Induced Pluripotent Stem Cells Market to Witness CAGR 8.7% Growth in Revenue During the Period 2026 – Cole of Duty

The healthcare industry has been focusing on excessive research and development in the last couple of decades to ensure that the need to address issues related to the availability of drugs and treatments for certain chronic diseases is effectively met. Healthcare researchers and scientists at the Li Ka Shing Faculty of Medicine of the Hong Kong University have successfully demonstrated the utilization of human induced pluripotent stem cells or hiPSCs from the skin cells of the patient for testing therapeutic drugs.

The success of this research suggests that scientists have crossed one more hurdle towards using stem cells in precision medicine for the treatment of patients suffering from sporadic hereditary diseases. iPSCs are the new generation approach towards the prevention and treatment of diseases that takes into account patients on an individual basis considering their genetic makeup, lifestyle, and environment. Along with the capacity to transform into different body cell types and same genetic composition of the donors, hiPSCs have surfaced as a promising cell source to screen and test drugs.

Report Highlights:

Get Sample Copy of Report @ https://www.persistencemarketresearch.com/samples/17968

Company Profile

Get To Know Methodology of Report @ https://www.persistencemarketresearch.com/methodology/17968

In the present research, hiPSC was synthesized from patients suffering from a rare form of hereditary cardiomyopathy owing to the mutations in Lamin A/C related cardiomyopathy in their distinct families. The affected individuals suffer from sudden death, stroke, and heart failure at a very young age. As on date, there is no exact treatment available for this condition.

This team in Hong Kong tested a drug named PTC124 to suppress specific genetic mutations in other genetic diseases into the iPSC transformed heart muscle cells. While this technology is being considered as a breakthrough in clinical stem cell research, the team at Hong Kong University is collaborating with drug companies regarding its clinical application.

The unique properties of iPS cells provides extensive potential to several biopharmaceutical applications. iPSCs are also used in toxicology testing, high throughput, disease modeling, and target identification. This type of stem cell has the potential to transform drug discovery by offering physiologically relevant cells for tool discovery, compound identification, and target validation.

A new report by Persistence Market Research (PMR) states that the globalinduced pluripotent stem or iPS cell marketis expected to witness a strong CAGR of 7.0% from 2018 to 2026. In 2017, the market was worth US$ 1,254.0 Mn and is expected to reach US$ 2,299.5 Mn by the end of the forecast period in 2026.

Access Full Report @ https://www.persistencemarketresearch.com/checkout/17968

Customization to be the Key Focus of Market Players

Due to the evolving needs of the research community, the demand for specialized cell lines have increased to a certain point where most vendors offering these products cannot depend solely on sales from catalog products. The quality of the products and lead time can determine the choices while requesting custom solutions at the same time. Companies usually focus on establishing a strong distribution network for enabling products to reach customers from the manufacturing units in a short time period.

Entry of Multiple Small Players to be Witnessed in the Coming Years

Several leading players have their presence in the global market; however, many specialized products and services are provided by small and regional vendors. By targeting their marketing strategies towards research institutes and small biotechnology companies, these new players have swiftly established their presence in the market.

See the article here:
Induced Pluripotent Stem Cells Market to Witness CAGR 8.7% Growth in Revenue During the Period 2026 - Cole of Duty

EMA warns against unproven therapies – English – Agenzia ANSA

(ANSA) - Rome, April 29 - The European Medicines Agency (EMA)has sounded the alarm about the use of cell-based therapies thatare promoted as being miracle cures but, in fact, are oftenunproven and unauthorized.Serious Risks. Some health facilities are offering these therapies in Europevia advertisements on the Internet. Patients desperately looking for cures for a variety ofillnesses are often lured to them, but "these treatments canpose serious risks to patients for little or no benefit" warnedthe EMA's Committee for Advanced Therapies (CAT). The committee said it has drafted a document in response to"individuals, companies and hospitals promoting unprovencell-based therapies as cures for a broad range of conditionsincluding cancer, cardiovascular diseases, autism, cerebralpalsy, muscular dystrophy and vision loss".Growing Phenomenon. Francesca Ceradini, the director of the Osservatorio TerapieAvanzate (Advanced Therapies Observatory), said that this was "agrowing phenomenon that is increasingly within reach. "Before, the journeys of hope (for cures) used to be to Indiaor China, but today the destinations are in Europe and theUnited States too," she said. Source of Hope. Cell-based therapies are treatments using cells from thepatient or a donor. These are used to regenerate tissue or organs and thesetechniques are also a source of hope for those examiningpossible treatments for COVID-19. The cells are manipulated in a laboratory (cultivated),genetically modified, or used for a different essential functionto the original one. They are regulated in the EU as medicinal products and theEMA's Committee for Advanced Therapies works to ensure "timelyaccess to these potentially life-changing treatments".Web Adverts. Alessandro Aiuti, the Deputy Director of the TIGET genetictherapy institute and a CAT member who was involved in draftingthe document, told ANSA that the EMA's concern stemmed fromadverts on the Internet. "We have received reports of adverts on the websites ofclinics in several European Union countries that offertreatments sold as miracle cures based on mesenchymal cells,wrongly called stem cells, for example, for the treatment ofAlzheimer's, with no scientific basis and with no proof ofeffectiveness," he said. "This takes us backwards to the mistakes made with the(discredited) Stamina (therapy)." Few Approved Cell Therapies. Ceradini said that, at the moment, there are very fewapproved cellular therapies in Europe. "Many are being tested and the rest amounts to a jungle ofunproven treatments," she said. "In the USA alone there are 700 private clinics that sellthem at a very high price. "But there are others in Europe, especially in the east, inSwitzerland and perhaps in Italy too". Side Effects Can Be Fatal. The EMA's statement said that patients using unproven orunregulated cell-based therapies "have reportedly sufferedserious, sometimes fatal, side effects including infections,unwanted immune reactions, tumour formation, loss of vision andbleeding in the brain". The EMA said that, in order to protect the public, "welldesigned clinical trials on the safety and benefits ofcell-based therapies are essential. "Patients or their families who are considering cell-basedtherapies should ask their healthcare professional about thebenefits and risks of the treatment and which authority hasapproved it".

Follow this link:
EMA warns against unproven therapies - English - Agenzia ANSA

Exosomes: Definition, Function and Use in Therapy – Technology Networks

What are exosomes?

Exosomes are a class of cell-derived extracellular vesicles of endosomal origin, and are typically 30-150 nm in diameter the smallest type of extracellular vesicle.1 Enveloped by a lipid bilayer, exosomes are released into the extracellular environment containing a complex cargo of contents derived from the original cell, including proteins, lipids, mRNA, miRNA and DNA.2 Exosomes are defined by how they are formed through the fusion and exocytosis of multivesicular bodies into the extracellular space.

Multivesicular bodies* are unique organelles in the endocytic pathway that function as intermediates between early and late endosomes.3 The main function of multivesicular bodies is to separate components that will be recycled elsewhere from those that will be degraded by lysosomes.4 The vesicles that accumulate within multivesicular bodies are categorized as intraluminal vesicles while inside the cytoplasm and exosomes when released from the cell.

*Confusingly, there is inconsistency in the literature; while some sources differentiate multivesicular bodies from late endosomes, others use the two interchangeably.

Exosomes are of general interest for their role in cell biology, and for their potential therapeutic and diagnostic applications. It was originally thought that exosomes were simply cellular waste products, however their function is now known to extend beyond waste removal. Exosomes represent a novel mode of cell communication and contribute to a spectrum of biological processes in health and disease.2One of the main mechanisms by which exosomes are thought to exert their effects is via the transfer of exosome-associated RNA to recipient cells, where they influence protein machinery. There is growing evidence to support this, such as the identification of intact and functional exosomal RNA in recipient cells and certain RNA-binding proteins have been identified as likely players in the transfer of RNA to target cells.5,6 MicroRNAs and long noncoding RNAs are shuttled by exosomes and alter gene expression while proteins (e.g. heat shock proteins, cytoskeletal proteins, adhesion molecules, membrane transporter and fusion proteins) can directly affect target cells.7,8Exosomes have been described as messengers of both health and disease. While they are essential for normal physiological conditions, they also act to potentiate cellular stress and damage under disease states.2

Multivesicular bodies are a specialized subset of endosomes that contain membrane-bound intraluminal vesicles. Intraluminal vesicles are essentially the precursors of exosomes, and form by budding into the lumen of the multivesicular body. Most intraluminal vesicles fuse with lysosomes for subsequent degradation, while others are released into the extracellular space.9,10 The intraluminal vesicles that are secreted into the extracellular space become exosomes. This release occurs when the multivesicular body fuses with the plasma membrane.

The formation and degradation of exosomes.

This is an active area of research and it is not yet known how exosome release is regulated. However, recent advances in imaging protocols may allow exosome release events to be visualized at high spatiotemporal resolution.11

Exosomes have been implicated in a diverse range of conditions including neurodegenerative diseases, cancer, liver disease and heart failure. Like other microvesicles, the function of exosomes likely depends on the cargo they carry, which is dependent on the cell type in which they were produced.12 Researchers have studied exosomes in disease through a range of approaches, including:

In cancer, exosomes have multiple roles in metastatic spread, drug resistance and angiogenesis. Specifically, exosomes can alter the extracellular matrix to create space for migrating tumor cells.13,14 Several studies also indicate that exosomes can increase the migration, invasion and secretion of cancer cells by influencing genes involved with tumor suppression and extracellular matrix degradation.15,16Through general cell crosstalk, exosomal miRNA and lncRNA affect the progression of lung diseases including chronic obstructive pulmonary disease (COPD), asthma, tuberculosis and interstitial lung diseases. Stressors such as oxidant exposure can influence the secretion and cargo of exosomes, which in turn affect inflammatory reactions.17 Altered exosomal profiles in diseased states also imply a role for exosomes in many other conditions such as in neurodegenerative diseases and mental disorders.18,19Cells exposed to bacteria release exosomes which act like decoys to toxins, suggesting a protective effect during infection.20 In neuronal circuit development, and in many other systems, exosomal signaling is likely to be a sum of overlapping and sometimes opposing signaling networks.21

Exosomes can function as potential biomarkers, as their contents are molecular signatures of their originating cells. Due to the lipid bilayer, exosomal contents are relatively stable and protected against external proteases and other enzymes, making them attractive diagnostic tools. There are increasing reports that profiles of exosomal miRNA and lncRNA differ in patients with certain pathologies, compared with those of healthy people.17 Consequently, exosome-based diagnostic tests are being pursued for the early detection of cancer, diabetes and other diseases.22,23Many exosomal proteins, nucleic acids and lipids are being explored as potential clinically relevant biomarkers.24 Phosphorylation proteins are promising biomarkers that can be separated from exosomal samples even after five years in the freezer25, while exosomal microRNA also appears to be highly stable.26 Exosomes are also highly accessible as they are present in a wide array of biofluids (including blood, urine, saliva, tears, ascites, semen, colostrum, breast milk, amniotic fluid and cerebrospinal fluid), creating many opportunities for liquid biopsies.

Exosomes are being pursued for use in an array of potential therapeutic applications. While externally modified vesicles suffer from toxicity and rapid clearance, membranes of naturally occurring vesicles are better tolerated, offering low immunogenicity and a high resilience in extracellular fluid.27 These naturally-equipped nanovesicles could be therapeutically targeted or engineered as drug delivery systems.

Exosomes bear surface molecules that allow them to be targeted to recipient cells, where they deliver their payload. This could be used to target them to diseased tissues or organs.27 Exosomes may cross the blood-brain barrier, at least under certain conditions28 and could be used to deliver an array of therapies including small molecules, RNA therapies, proteins, viral gene therapy and CRISPR gene-editing.

Different approaches to creating drug-loaded exosomes include27:

Exosomes hold huge potential as a way to complement chimeric antigen receptor T (CAR-T) cells in attacking cancer cells. CAR exosomes, which are released from CAR-T cells, carry CAR on their surface and express a high level of cytotoxic molecules and inhibit tumor growth.29 Cancer cell-derived exosomes carrying associated antigens have also been shown to recruit an antitumor immune response.30

The purification of exosomes is a key challenge in the development of translational tools. Exosomes must be differentiated from other distinct populations of extracellular vesicles, such as microvesicles (which shed from the plasma membrane, also referred to as ectosomes or shedding vesicles) and apoptotic bodies.31 Although ultracentrifugation is regarded as the gold standard for exosome isolation, it has many disadvantages and alternative methods for exosome isolation are currently being sought. Exosome isolation is an active area of research (see Table 1) and many research groups are seeking ways to overcome the disadvantages listed below, while navigating the relevant regulatory hurdles along the way.

Produces a low yield and low purity of the isolated exosomes as other types of extracellular vesicles have similar sedimentation properties.

Low efficiency as it is labor-intensive, time-consuming and requires a large amount of sample. specialized equipment. High centrifugal force can damage exosome integrity

See the article here:
Exosomes: Definition, Function and Use in Therapy - Technology Networks

Remestemcel-L Looks Promising for COVID-19 With Moderate to Severe ARDS – Pulmonology Advisor

Home Topics Lung Infection

Mesoblast announced data from a phase 2/3 trial evaluating remestemcel-L, an allogeneic mesenchymal stem cell product candidate, in ventilator-dependent COVID-19 patients with moderate to severe acute respiratory distress syndrome (ARDS).

Remestemcel-L consists of culture-expanded mesenchymal stem cells derived from the bone marrow of an unrelated donor. It is believed to work by down-regulating the production of proinflammatory cytokines, increasing production of anti-inflammatory cytokines, and enabling recruitment of naturally occurring anti-inflammatory cells to involved tissues.

The randomized, placebo-controlled trial is being conducted at Mount Sinai hospital in New York City. Patients were treated with a variety of experimental agents prior to receiving remestemcel-L. Findings from the study showed 83% survival in ventilator-dependent COVID-19 patients with moderate/severe ARDS (n=10/12) following 2 intravenous infusions of remestemcel-L within the first 5 days; 75% of patients (n=9/12) were able to successfully come off ventilator support at a median of 10 days. There have been 7 patients discharged from the hospital as of now.

Mesoblast Chief Executive Dr. Silviu Itescu stated: The remarkable clinical outcomes in these critically ill patients continue to underscore the potential benefits of remestemcel-L as an anti-inflammatory agent in cytokine release syndromes associated with high mortality, including acute graft versus host disease and COVID-19 ARDS. We intend to rapidly complete the randomized, placebo-controlled phase 2/3 trial in COVID-19 ARDS patients to rigorously confirm that remestemcel-L improves survival in these critically ill patients.

Additionally, the Food and Drug Administration recently accepted for Priority Review the Biologics License Application of remestemcel-L for the treatment of steroid-refractory acute graft vs host disease. The Company expects to launch remestemcel-L in 2020 if approved.

For more information mesoblast.com.

This article originally appeared on MPR

Please login or register first to view this content.

LoginRegister

Next post in Lung InfectionClose

Read more here:
Remestemcel-L Looks Promising for COVID-19 With Moderate to Severe ARDS - Pulmonology Advisor

First COVID-19 Convalescent Plasma DonorCanadian Blood Services begins collecting plasma donations from people who have recovered from COVID-19 as…

VANCOUVER, British Columbia, April 29, 2020 (GLOBE NEWSWIRE) -- Canadian Blood Services is proud to be part of CONCOR, a national clinical trial to test the safety and effectiveness of COVID-19 convalescent plasma as a possible treatment to help patients infected with the virus. Today, the national blood authority and operator collected its first COVID-19 convalescent plasma donation in Vancouver.

Im delighted to be able to help out a really good cause. I have been an active blood donor for 15 years. Im pleased my unfortunate situation can help somebody else. I think Im doing a little bit of good out of all this. says Jerry Glubisz, Canadian Blood Services first COVID-19 convalescent plasma donor.

Canadian Blood Services is establishing a national convalescent plasma collection program. The blood operator is now recruiting potential convalescent plasma donors across the country through its new online registry.

Convalescent plasma may help patients recover from COVID-19, but this has not yet been proven. Well-designed clinical trials, like CONCOR, will help provide the necessary information about whether this is a safe and effective treatment option for patients, says Dr. Dana Devine, chief scientist with Canadian Blood Services. Were making an important contribution to research on a global scale that could help patients in Canada and around the world.

Over the next few weeks more convalescent plasma donors, like Jerry Glubisz, may donate at one of 11 Canadian Blood Services donor centres that have the capability to collect blood components, like plasma, through a process called apheresis. These donor centres are located in Vancouver, Calgary, Edmonton, Saskatoon, Regina, Winnipeg, London, Hamilton, Toronto, Ottawa and Halifax.

Initially, all convalescent plasma donations will be supplied to Canadian physicians caring for patients with COVID-19 in the context of the CONCOR trial and under the authorization of Health Canada.

Including both of Canadas public blood operators (Canadian Blood Services and Hma-Qubec), there are 10 research teams and more than 50 hospitals across the country currently participating in CONCOR. Patient involvement in the clinical trial will be determined by their treating physician at a participating hospital in consultation with the patient and/or the patients family. Patients with COVID-19 who are looking for more information on convalescent plasma as a treatment option are encouraged to visit CONCOR1.ca.

While Canadian Blood Services has officially started collecting convalescent plasma, these donations must still undergo all necessary testing and processing before being issued for use by physicians, as per blood safety and quality standards. Transfusions of convalescent plasma are expected to begin within a few weeks when the trial begins.

A convalescent plasma donation is the same as a plasma donation; however, a specific donor is needed for this clinical trial. In addition to meeting Canadas current plasma donor eligibility criteria, convalescent plasma donors must be younger than 67 years of age, previously confirmed positive for COVID-19 by a laboratory test, and fully recovered from the virus and symptom free for at least 28 days to participate. Donors must also live within driving distance of a donor centre located in one of the aforementioned cities. Anyone who meets these requirements is encouraged to join Canadian Blood Services online registry. Additional testing will be done at the time of collection to ensure there are adequate antibodies against the COVID-19 virus in the donors plasma to be part of the trial.

Registered convalescent plasma donors who may be eligible are being contacted now and all convalescent plasma donation appointments are being booked as donors are qualified to participate by Canadian Blood Services Centre for Innovation which oversees research and development for the organization.

Canadian Blood Services is not accepting walk-in donors for any of its collection programs during the pandemic.

Media assets

As per current physical distancing measures, media are not permitted to visit Canadian Blood Services donor centres. To ensure media have access to assets necessary to provide coverage, raw footage and an uncut interview with Canadian Blood Services first convalescent plasma donor will be made available.

Web-based interviews with Mr. Glubisz and with convalescent plasma donors in other areas of Canada may also be arranged through Canadian Blood Services.

Please contact media@blood.ca for more information.

About Canadian Blood Services

Canadian Blood Services is a not-for-profit charitable organization. Regulated by Health Canada as a biologics manufacturer and primarily funded by the provincial and territorial ministries of health, Canadian Blood Services operates with a national scope, infrastructure and governance that make it unique within Canadian healthcare. In the domain of blood, plasma and stem cells, we provide services for patients on behalf of all provincial and territorial governments except Quebec. The national transplant registry for interprovincial organ sharing and related programs reaches into all provinces and territories, as a biological lifeline for Canadians.

About Canadian Blood Services Research Activities

Canadian Blood Services, through its Centre for Innovation, conducts and supports research projects in key priority areas that span the translational continuum from bench to bed side. The focus is on transfusion science and medicine but also related fields such as cellular therapies (in particular hematopoietic stem cell transplantations) and organ and tissue transplantation. Our research findings are published in peer-reviewed journals or directly shared with stakeholders.

Instrumental to our research efforts are discovery and applied research laboratories led by university-affiliated Canadian Blood Services staff scientists, as well as our medical experts and adjunct scientists. Complementing these core research teams, the Centre for Innovation facilitates a national and international research network of blood system experts through competitive research funding opportunities, collaborations and contract research.

/CanadasLifeline@CanadasLifeline1 888 2DONATE

FOR MORE INFORMATIONTEL. 1-877-709-7773EMAILmedia@blood.ca

Read the original post:
First COVID-19 Convalescent Plasma DonorCanadian Blood Services begins collecting plasma donations from people who have recovered from COVID-19 as...

Cancer Stem Cell Therapies: Mapping the Future Growth Potential – Communal News

The objective of the ongoing R&D in this domain is to develop novel CSC directed treatments that can combat complications, such as off-target toxicities and disease relapse, associated with the current standard of care treatments.TheCancer Stem Cell Therapies Market, 2017-2030report examines the current landscape and the future outlook of the growing pipeline of products targeting CSCs.Some of the most interesting insights from our study are presented below.

CSCs have Garnered a Lot of Interest Across Different PlatformsIncreasing chatter on social media, and the fact that over 9,000 scientific articles have been published in the last few years, indicate the growing interest in this domain

A Robust Pipeline Offers Significant Future PotentialWith two commercialized drugs and close to 150 clinical/preclinical molecules, the field has evolved significantly over time and has several promising candidate therapies

A Healthy Mix of Small Molecules and Biologics, The growing pipeline features both small molecule and biologic product candidates, which are being developed to target a number of cancer stem cell regulatory pathways

Being Designed for Novel Targets, Reinforces the Underlying PromiseSpecifically, over 70 clinical-stage molecules are being investigated to tap opportunities across different types of cancer

The US and the EU are Key Innovation HubsBoth big and small pharma players are actively involved; the US and EU are currently leading the research efforts

The Market is Expected to Witness Rapid Growth in the Mid-Long TermAfter the approval and launch of multiple late-phase product candidates, the market is expected to grow at a rapid pace

The Opportunity is Likely to be Well Distributed Across Indications and TargetsThe emergence of effective therapeutic strategies will result in better opportunities in the long term as more drugs get approved for a diverse range of indications

To know more please clickhere For any queries/suggestions or if youd like to get access to the full research results, please do not hesitate to contact us atsales@rootsanalysis.com

Continue reading here:
Cancer Stem Cell Therapies: Mapping the Future Growth Potential - Communal News