Emerging potential of PTL nanoformulations in tumor therapy | DDDT – Dove Medical Press

Introduction

As a medical herb, Feverfew (Tanacetum parthenium) is conventionally used in Europe to treat fever, inflammation, migraines, rheumatoid arthritis, and menstrual irregularities. Parthenolide (PTL, Figure 1) is a sesquiterpene lactone found in feverfew, which is currently considered to be responsible for the herbs therapeutical potential.1 Initially, conventional extraction methods using chloroform and petroleum ether were performed to extract PTL; various extraction methods have been developed, such as high-performance liquid chromatography (HPLC), Soxhlet extraction, supercritical fluid extraction (SFE) and microwave-assisted extraction (MAE).1,2

Figure 1 Chemical structure of PTL and DMAPT.

The anti-cancer property of PTL was firstly validated in 1973. Furthermore, its patent application for tumor suppression was approved in 2005.3 Additionally, the in vitro and in vivo antitumor potential of PTL in multiple cancer types has been confirmed by numerous researches, which mainly resulted from its cytotoxicity to the bulk population of cancer cells as well as from selectively targeting cancer stem cells (CSCs); it is a subpopulation currently believed to be responsible for chemotherapy resistance and tumor relapse.310 Further studies revealed a series of direct PTL targets [p65, IB kinase (IKK), focal adhesion kinase 1 (FAK1), and others] that indirectly affect signaling pathways, which account for cell cycle arrest, apoptosis induction, metastasis suppression, redox imbalance, and epigenetic regulation involved in PTLs antitumor properties.3,4,11,12 The potential utility of PTL as radio-sensitization agent and complementary therapy against various cancers has also been widely studied and summarized.1315 As reviewed by Malgorzata et al, PTL has been combined with various anticancer agents, such as tubulin-directed agents, anthracyclines, antimetabolites, histone deacetylase inhibitors, mTOR inhibitors, and inducers of reactive oxygen species (ROS).14

Despite its deciphered anticancer potential and mechanisms of action in pre-clinical experiments, the clinical application of PTL remains hindered because of some disadvantages, including weak aqueous solubility, low oral bioavailability, and relative instability under chemical and physiological conditions.16,17 As a result, various methods for synthesizing PTL derivatives to yield compounds with better hydrophilicity and improved potency have been proposed.4,6,18 Dimethylamino parthenolide (DMAPT, Figure 1), a representative among hydrophilic PTL analogues, showed improved water solubility and oral bioavailability. Thus, it has advanced into the first phase of a clinical trial for the treatment of acute myeloid leukemia (AML).19

Nanomedicine is a rapidly developing field that exploits nanoparticles (NPs) to facilitate the diagnosis and treatment of a wide range of diseases. Nanoparticles applied in nanomedicine generally refer to a type of colloidal drug delivery system, which comprises particles with a size range from 10 to 200 nm in diameter.20 By far, diverse types of nanoparticles have been developed as drug carriers, including but not limited to liposomes, polymeric micelles, carbon nanotubes, mesoporous silica nanoparticles, metal-based nanoparticles, and dendrimers. Moreover, these can be made of diverse materials, including lipids, phospholipids, polymers, proteins, and inorganic materials.21,22

Compared with free drug counterparts, nanoparticles entrapment has displayed distinct advantages, such as improved bioavailability, prolonged circulation time, and ease of functionalization by surface modification. Furthermore, the enhanced permeability and retention (EPR) effect caused by the large amount of leaky vascularization and impaired lymphatic drainage at the tumor site enables non-targeted nanoparticles to accumulate in tumor tissues.23 Some of these nanoparticles have been approved as cancer therapeutics by the Food and Drug Administration.21 Besides the chemical modification of PTL for property improvement, the development of nanoscale drug delivery systems offers another promising strategy to overcome the poor water solubility and bioavailability of PTL as well as to determine its efficient and selective delivery to tumor tissues; the latter of which has not been summarized as compared to the extensively reviewed bioactivities and combination treatments of PTL. Therefore, we focused on the key antitumor mechanisms of PTL as well as its efficiency in being formulated as a nanoparticle delivery system.

Current evidence demonstrates that the antitumor mechanism of PTL is multifactorial and complex, due to the high electrophilic reactivity of -methylene--lactone present in PTL, thereby resulting in the alkylation of various proteins. A number of PTL targets have thus been identified and summarized.4 Furthermore, newer potential targets of PTL, such as USP7 and EGFR, continue to be reported,2427 thereby giving rise to the regulatory effect of PTL on various signaling pathways (Figure 2) and cellular processes (Figure 3) responsible for proliferation, cell cycle regulation, stemness, cell death, angiogenesis and metastasis.3,4 Thus, it is not surprising that PTL displays diverse anticancer effects, including abrogated cell viability and angiogenesis, cell cycle arrest, cell death induction, and decrease in stemness, invasiveness, and chemoresistance (Table 1).

Figure 2 Selected signaling pathways regulated by PTL. The key components and signal transduction cascade reactions in NF-B signaling, Wnt/-catenin signaling, JAK/STAT signaling, FAK1 signaling, and p53 signaling were depicted, and the nodes targeted by PTL were labeled.

Notes: Represents the inhibitory effect, and Represents the activation effect.

Figure 3 Pivotal cellular processes affected by PTL. Schematic diagram showing the formation of microtentacle and major antioxidant machineries were plotted. The targets regulated by PTL were marked.

Note: Represents the accumulation of ROS.

As shown in Figure 2, several signaling pathways closely related to tumorigenesis and progression were suppressed by PTL; among these, the prominent NF-B signaling pathway was the first to be inhibited. Mechanism investigation illustrated that PTL inhibited NF-B signaling by alkylating cysteine 38 in p65 and cysteine 179 in IKK.28,29 Additionally, a later study predicted that tumor necrosis factor receptor-associated factor 6 (TRAF6) might have been a novel target involved in the PTL-associated inhibition of NF-B.30 Moreover, the role of PTL in preventing NF-B activation contributed to the suppression of hypoxia-inducible factor-1 (HIF-1) signaling.31 Recent studies have elucidated that PTL inhibited Wnt/-catenin signaling by targeting ubiquitin specific peptidase 7 (USP7) and ribosomal protein L10 (RPL10), a deubiquitination enzyme stabilizing -catenin and a ribosomal protein related to the synthesis of the transcriptional regulator 4/lymphoid enhancer binding factor 1 (TCF4/LEF1), respectively.24,32 Furthermore, FAK1 and janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling were found to be impaired by PTL by covalently modifying specific cysteine molecules.33,34

Apart from inhibiting the aforementioned signaling pathways, PTL also activated p53 functions by promoting the ubiquitination of E3 ligase murine double minute 2 (MDM2) in an ataxia-telangiectasia mutated (ATM)-dependent manner, thus leading to either cell cycle arrest or apoptosis.35 Specially, USP7 was reported to interact with and stabilize MDM2 and p65.36,37 Thus, the inhibitory effect of PTL on USP7 might be a further step for activating p53 and inhibiting NF-B. Additionally, the activation of c-Jun N-terminal kinase (JNK) by PTL enhanced the sensitivity of human cancer cells to tumor necrosis factor- (TNF-) or tumor necrosis factor-related apoptosis-inducing ligand (TRAIL).38,39

To date, PTL has been found to exert antitumor effects on several cellular processes, including redox balance and epigenetic modulation; the targets involved in these two processes have also been revealed (Figure 3). Several studies have indicated that PTL disrupted intracellular redox homeostasis by depleting glutathione (GSH) and inhibiting its metabolic enzymes, including glutamate-cysteine ligase catalytic subunit (GCLC), thioredoxin reductase 1/2 (TrxR1/2), and GSH peroxidase 1 (GPX1), thus leading to increased ROS level.4042 Moreover, ROS enhancement by PTL seemed to elicit different forms of cell death, such as necrosis, apoptosis, and autophagic cell death, depending on the tumor cell type.40,41,43 Meanwhile, the action of PTL on ROS partially accounts for its distinctive ability to selectively induce cell death in cancer cells, while sparing the equivalent normal cells. This is due to the fact that oxidative stress in cancer cells is more frequently elevated than that in normal cells. Thus, additional ROS produced by PTL may promote tumor cell death, whereas normal cells may maintain redox homeostasis by adaptive antioxidant responses.44 Moreover, the epigenetic targets of PTL consist of various enzymes, including histone deacetylase 1 (HDAC1), DNA methyltransferase 1 (DNMT1), and lysine methyltransferase 5C (KMT5C); these regulate the transcription of various genes, such as p21 and high in normal-1 (HIN-1).4547 In addition, PTL disrupted the cyclical detyrosination/tyrosination of tubulin by inhibiting tubulin carboxypeptidase (TCP), which led to a reduced frequency of microtentacles and suppressed tumor cell reattachment to endothelial layers (Figure 2).48,49

Interactions between signaling pathways and cellular processes may be affected by PTL. For instance, the stimulated generation of ROS by PTL was able to induce autophagy and cause NF-kB downregulation.40,43 Furthermore, NF-B inhibition contributed to epigenetic regulation,47 thereby further complicating the antitumor mechanism of PTL.

To understand the pharmacological effects of PTL, in vitro studies on its efficacy, pharmacological activities, and potential molecular mechanisms in a variety of cancer cells as well as available in vivo models since 2019 are tabulated in Table 1, thus further extending and qualifying existing reviews of its biological activities.3,4,50 As shown in Table 1, PTL modulation toward the aforementioned signaling pathways and cellular processes exerts multiple pharmacological effects against a myriad of tumor cell types.

The antiproliferative activity of PTL was detected in almost all studies; the results indicated that PTL exhibited half-maximal inhibitory concentration (IC50) between the range of 2.525 M for most tumor cells listed, thus showcasing its cytotoxicity to different cancer cells.5159 Cell cycle arrest, induction of cell death, and changes in related proteins were further detected, thus further supporting the cytotoxic potential of PTL. Overall, PTL was shown to induce different effects depending on the cell type, which can be illustrated by its ability to induce cell cycle arrest at different phases as well as several types of cell death in various cancer cells. For example, PTL arrested uveal melanoma cells in the G1 phase by upregulating p21 and downregulating cyclin D1, which are two G1 phase cell cycle regulatory proteins.60 On the other hand, PTL decreased the expression of survivin promoting G2/M cell cycle transition, thereby triggering G2/M cell cycle arrest in glioblastoma cells.61 Apoptosis, necrosis, and autophagy are the three forms of cell death caused by PTL;62 among which, apoptosis is the most studied. It is well known that apoptosis is elicited by two distinct pathways, the extrinsic and mitochondria-mediated intrinsic pathways; it culminates in the activation of caspases, which function as the main apoptotic effectors.63

Numerous studies demonstrated that PTL treatment could induce extrinsic or intrinsic apoptosis in tumor cells by inhibiting the activities of the NF-B, STAT3, Wnt and JNK signaling pathways, activating the p53 signaling pathway, regulating the Bcl-2 family members, and generating ROS.24,62,6471 Furthermore, PTL-induced cell death of breast cancer and multiple myeloma cells was dramatically attenuated by co-treatment with the pan-caspase inhibitor, Q-VD-OPh or Z-VAD-fmk, thus indicating that caspases are involved in PTL-induced apoptotic cell death; it also concurs the presence of other forms of cell death.33,72 Indeed, a large number of studies reported that PTL was capable of inducing autophagic or necrotic cell death.50,62 For instance, PTL mediated cell death through ROS-mediated autophagy in human osteosarcoma (Saos-2 and MG-63) and triple-negative breast cancer (MDA-MB-231) cells.40,43 In particular, PTL was capable of simultaneously inducing mixed forms of cell death, as evidenced by observations of PTL-induced apoptosis and autophagy in Hela and HepG2 cells as well as apoptosis and necrosis in HL60 cells.7375 Additionally, the role of autophagy in cell death involves the fact that its inhibition significantly blocked PTL-induced apoptosis in pancreatic cancer cells,76 but potentiated PTL-induced apoptosis in human breast cancer cells.77

Although previous studies have elucidated the selective targeting effect of PTL on CSCs from primary or sensitive cancer cells,3 a recent study by Yi et al suggested that PTL also effectively eliminated leukemia stem cells (LSCs) from adriamycin (ADM)-resistant K562 cells (K562/ADM) by suppressing aberrantly activated NF-B.64 In addition, NF-B inactivation by PTL sensitized gastric tumor and esophageal squamous cell carcinoma (ESCC) cells to chemotherapeutic drugs including ADM and cisplatin.78,79 PTL exerted its anti-angiogenic effects by inhibiting the NF-B-mediated VEGF expression in ESCC cells.79 Moreover, several studies have pointed out that PTL inhibits migration, invasion, and metastasis, which benefits from its abilities to regulate epithelial-to-mesenchymal transition (EMT) and to inhibit FAK1, TCP, and STAT3.33,8083

In short, the ultimate outcomes resulting from the affected signaling pathways and cellular processes by PTL include, but are not limited to, impaired cell proliferation and angiogenesis, induction of cell death, and reduced stemness, invasiveness, and chemoresistance. This has been confirmed by a large body of research, thus providing a sufficient basis for studies investigating and developing a wide range of PTL nanoformulations for various cancer therapies.

To the best of our knowledge, several types of PTL nanocarriers, including liposomes,8489 polymeric micelles,9096 nanocrystals,97 PLGA nanoparticles,98 and nanographene,99 have been synthesized to deliver PTL and ameliorate its anti-cancer efficacy. Whether PTL is co-encapsulated into nanoparticles with other drugs or small molecules and whether the nanoparticles are modified for targeted therapy, these studies can be classified into three types. Furthermore, the types, materials, and properties of nanocarriers mentioned in these studies, as well as the in vitro and in vivo models employed to evaluate the anticancer effects of nanoformulations, are separately tabulated in Tables 24.

Table 2 Unmodified Nanoparticles Solely Incorporated with PTL

Table 3 Undecorated Nanovectors Encapsulated PTL and Other Agents

Table 4 Targeted Nanocarriers Encapsulated PTL with or without Other Agents

Baranello et al synthesized different types of micelles formed from amphiphilic diblock copolymers of PSMA-b-PS or PSMA-b-PBA; PTL was successfully loaded into these micelles. However, PTL exhibited the greatest loading efficiency and capacity in PSMA100-b-PS258 micelles, thereby indicating that the composition and hydrophobic core chemistry of micelles were significant parameters for optimization.90 Although subsequent studies suggested that PTL-loaded PSMA100-b-PS258 micelles did not exhibit better cytotoxic ability toward MV4-11 cells than free PTL, it protected sequestered PTL from damage by both cells and deactivating chemicals, such as GSH.91 Similarly, the application of stealthy liposomes and micelles fabricated by F127 or biodegradable PTL-PTMC as nanocarriers of PTL did not appear to efficiently increase its cytotoxicity against MCF-7, MDA-MB-231, and patient T-lineage acute lymphoblastic leukemia (T-ALL) or B-cell precursor acute lymphoblastic leukemia (BCP-ALL) cells.89,92,93 However, the combination of stealthy liposomal PTL slightly sensitized the antitumor effects of stealthy liposomes loaded with vinorelbine.89

Unlike the nanocarriers mentioned above, the carboxyl-functionalized nanographene (fGn) as well as the PTL-nanocrystal delivery system showed improved antiproliferation activities in comparison with individual PTL. The IC50 of PTL and PTL-fGn for Panc-1 cells were 39 and 9.5 M respectively, whereas the IC50 of PTL for HepG2 cells decreased from 50.891 M to 33.618 M when delivered with nanocrystals.97,99 Interestingly, there was no significant difference in cytotoxic activity between DMAPT and DMAPT-fGn; this inefficacy may be due to the lack of a hydrophobic interaction between DMAPT and fGn.99 Thus, whether incorporation of DMAPT into other nanovectors could enhance its cytotoxicity is worth exploring. Furthermore, the in vitro and in vivo combination of PTL-nanocrystals and sorafenib achieved remarkable synergistic anti-cancer effects, as reflected by the MTT, wound-healing and HepG2 xenograft assays.97

Given the molecular complexity of cancer, combination therapy has attracted tremendous attention because of its ability to increase drug efficacy, improve drug resistance, and reduce systemic toxicity. Kanwaldeep et al constructed a paclitaxel and PTL co-delivery system of PEG2000-DSPE/vitamin E-TPGS mixed micelles, which retained a high encapsulation efficiency (>95%) and chemical stability over a storage period of 45 days. Furthermore, co-encapsulation of these two drugs significantly suppressed the viability of sensitive and taxol-resistant A549 cells compared to their free drug counterparts in solutions and single drug-loaded micelles.94 In addition, as a component of these mixed micelles, it is noteworthy that vitamin E-TPGS aided in not only maintaining high encapsulation efficiency due to its bulky structure and large surface area, but also enhanced chemosensitization by inhibiting P-glycoprotein (P-gp) efflux.94,100 Recently, a liposome system loaded with betulinic acid, PTL, honokiol and ginsenoside Rh2 displayed in vitro and in vivo antitumor activity comparable to cisplatin, the first-line therapy for lung cancer. In addition, this cocktail liposome therapy circumvented obvious kidney damage induced by cisplatin, as revealed by hematoxylin and eosin (H&E) staining. It also did not cause any significant damage to other major organs, including the heart, liver, spleen, and lungs, thereby indicating that this cocktail is a safer alternative for lung cancer treatment.84

To exploit the advances in photothermal therapy, Jin Xin and co-workers constructed thermosensitive liposomes (TSLs), in which PTL and the photosensitizer indocyanine green (ICG) were co-loaded. ICG converts light energy into heat energy upon near-infrared light irradiation. Compared to groups treated with paclitaxel, a combination of free drugs with or without laser, or PTL-ICG TSLs without laser, groups treated with PTL-ICG TSLs with laser exhibited lower cell viability, higher ROS induction and apoptosis, and better in vivo anti-cancer effects. According to these results, the two benefits of heat energy released by ICG under near-infrared radiation at the tumor location were validated. On the one hand, the heat-promoted phase transition of these liposomes enhanced their fluidity and permeability, thus allowing loaded drugs to effectively diffuse to tumor tissues at high concentrations. On the other hand, the maximum temperature of tumor tissues treated with PTL-ICG TSLs with laser reached 47.4C 2.68C, which led to irreversible damage and further synergized with the oxidative stress of PTL.85

To further improve drug delivery and selective targeting toward cancer cells, targeted nano-encapsulation of PTL with or without other agents was developed according to the characteristics of specific types of tumor cells and their microenvironment or the suction of the magnetic field. CD44 is a pivotal receptor involved in myelopoiesis; its specific variant isoforms have been reported to be overexpressed in AML cells, thus indicating that CD44 can serve as a promising receptor for targeted delivery of anti-AML drugs.101,102 As a result, an intervention was developed by encapsulating PTL into PLGA nanoparticles conjugated with antiCD44 with higher tumor targeting efficiency than PLGA-PTL nanoparticles. Although the cytotoxic abilities of PLGA-antiCD44-PTL and PLGA-PTL nanoparticles were not compared, PLGA-antiCD44-PTL nanoparticles exhibited stronger cytotoxicity than free PTL.98

Leukemia stem cells (LSCs) within the bone marrow (BM) microenvironment are thought to be the primary mediators of relapse and chemotherapy resistance in AML. Furthermore, E-selectin expressed in the BM endothelium provides a feasible approach for targeted BM delivery. In view of the remarkable capability of PTL to eradicate cancer stem cells including LSCs, a multistage vector system (MSV) was developed by entrapping PTL in mPEG-PLA micelles coated with protective degradable porous silicon particles and an E-selectin thioaptamer. In contrast to the negligible therapeutic efficacy of PTL-loaded micelles, MSV-PTL significantly reduced the tumor burden of patient-derived AML xenografts, concurrent with the inhibition of NF-B and activation of -H2AX; this supports the effectiveness of the MSV system for targeted BM delivery. Moreover, the decreased level of secondary xenotransplants implied that the directed delivery of PTL to the BM using the MSV system led to the elimination of LSCs.95

The tLyP-1 peptide has been verified to possess cell-penetrating ability and tumor-targeting capacity, which are derived from its C-terminal structure and affinity to the neuropilin-1 receptor overexpressed in several kinds of cancer cells (eg glioma and lung cancer).103 In which tLyP-1-modified liposomes entrapped in PTL and ginsenoside compound K (CK) were synthesized, the level of ROS and induced apoptosis of A549 cells in vitro significantly increased. Besides, PTL/CK tLyP-1 liposomes exhibited greater anticancer efficacy than the combined administration of these two compounds in A549 tumor-bearing mice.87 In addition, Ran et al developed PTL-loaded PEG-PLA micelles decorated with a Y-shaped DWVAP peptide, which could guide micelles across multiple biological barriers and ultimately target glioma and its associated stem cells. Moreover, combined therapy of PTL-loaded, DWVAP-modified PEG-PLA micelles with temozolomide or DWVAP-modified PEG-PLA micelles loaded with paclitaxel achieved outstanding anti-glioma efficacy, according to the KaplanMeier survival analysis.96

Recently, by virtue of the magnetic field, magnetic nanoparticles (MNPs) were chosen to modify the surface of liposomes (lips) loaded with PTL by Gao and co-workers.86,88 In one study, PTL-ICG-Lips, similar to PTL-ICG TSLs mentioned above, were coated with MNPs. In terms of in vitro heating efficiency and drug release, there was no significant difference between the [emailprotected] group treated only with laser and [emailprotected] group treated with laser plus magnet, which were stronger than non-laser irradiation treated groups. These results suggested the dominant role of photothermal conversion mediated by ICG in these processes. However, the presence of the magnetic field increased the heating rate and percentage of drug release at an earlier stage. In addition, cells treated with [emailprotected] (magnet plus laser) exhibited the highest cellular uptake. Consistently, in vivo studies also showed that the [emailprotected] (magnet plus laser) group displayed the fastest heating rate, highest temperature, and highest intratumoral PTL concentration.86 These findings indicated that the magnetic field could enrich the magnetic liposomes in the irradiated area, thus further enhancing the efficiency of photothermal conversion and facilitating the release and uptake of PTL.

Another multifunctional delivery system generated by Gao et al was the encapsulation of PTL and glucose oxidase (GOD) into nanomagnetic liposomes coated with chitosan, named [emailprotected] The addition of chitosan endowed [emailprotected] with the capability to release drug at a slightly acidic pH, which was characteristic of the tumor microenvironment. As a result, this increased the targeting ability of the system together with the magnetic field. By consuming glucose, GOD in this system lowers the pH and generates H2O2 as well as starves the cancer cells to death. A lower pH further promotes drug release; H2O2 can be subsequently catalyzed by iron ions in MNPs to produce hydroxyl radicals (OH), a noxious ROS. Meanwhile, PTL protects OH from scavenging by depleting GSH,40 which amplifies the intracellular oxidative stress and thus leads to cell apoptosis. Under an extra magnetic field, the [emailprotected] demonstrated prominent antitumor effects in vitro and in vivo through the integration of chemo-, chemodynamic, starvation and magnetic-targeting therapies.88

As discussed above, the incorporation of PTL into nanocarriers results in increased solubility, cellular uptake and stability, prolonged circulation time, and enhanced accumulation at tumor sites. Therefore, the majority of nanocarriers encapsulated with PTL, especially when combined with other agents such as photosensitizers, anticancer drugs, and MNPs, demonstrated higher anticancer efficiency than free PTL. These mechanisms are reflected by better antiproliferative activities, more effective induction of apoptosis, higher suppression rate of migration, and xenograft tumor growth. Histological examination demonstrated the low toxicity of this novel therapeutic agent.

In addition, a variety of naturally occurring sesquiterpene lactones structurally related to PTL with anticancer efficiency, as represented by micheliolide (MCL, Figure 4), melampomagnolide B (MMB, Figure 4), and costunolide (COS, Figure 4) have been reported.104106 Similar with PTL, most of current researches focus on structural modification of these compounds to improve their antitumor effects, stability, and sustainable release.107111 Several studies started to investigate the incorporation of MCL analogs and COS into nanoparticles.112,113 Bone-targeted PSMA-b-PS NPs entrapped with triazole MCL analog exhibited excellent serum stability and significantly reduced LSC burden in leukemic mice.112 Another study demonstrated that COS and COS-NPs, in combination with doxorubicin (DOX), stimulated the activity of caspase-3 and induced apoptosis of HCT116 and MDA-MB-231-Luc. They also suppressed the tumor growth of HCT116 and MDA-MB-231-Luc implants in nude mice. There was no significant difference in the anti-tumor activity of COS and COS-NPs; the authors deemed that this may have been due to the dose-selective approach for determining the optimal anti-cancer activity for both COS and Nano-COS. Thus, dose-response relationship will be investigated in future studies.113 In addition, Niu et al constructed pH-responsive mesoporous silica nanoparticles (MSNs) loaded with COS, which increased stability and enhanced anti-fibrotic effect of pure COS.114 In short, above-mentioned results implied that nanoparticles entrapped with sesquiterpene lactones show great promise in the treatment of cancer and other diseases.

Figure 4 Chemical structure of MCL, MMB and COS.

Studies from the past decades have validated the great potential of PTL as an anticancer agent with extremely intricate bioactivities. However, poor aqueous solubility, instability, low bioavailability, and drug-targeting property of PTL limit its in vivo anticancer efficacy and clinical application.86 As such, the development of nanoparticle-based platforms has been utilized in multiple biomedical fields, including hydrophobic drug delivery, which undoubtedly provides a promising strategic improvement.115 Indeed, several natural product-derived anticancer nanodrugs, including nanoparticle albuminbound (NAB)-paclitaxel and liposomal vincristine, have been used in clinical practice.21 However, no clinical trials have been reported for nanocomposites of PTL and its structurally related sesquiterpene lactones. Furthermore, despite multiple in vitro and in vivo experiments that have reported the benefits of nanoparticle-based formulations of other natural anti-cancer drugs in the treatment of various cancer types, including quercetin, curcumin, resveratrol, and andrographolide, only a few clinical trials have been performed, thus, suggesting that the investigation of these nanocarriers is still in its relative infancy.116118 Further optimization can be performed because the efficiency of nanoparticles can be influenced by many parameters, such as nanocarrier types, compositions (eg materials, ligand modification, co-encapsulated agents), and physical properties (eg size, shape, surface charge).21 As such, PTL and other natural product-based nanoformulations with improved properties will undoubtedly emerge; clinical trials need to be encouraged to further validate the security and therapeutic efficiency of these nanoparticles for cancer. Finally, PTL has been recently predicted to be a possible agent for the treatment of other diseases, such as Hutchinson-Gilford Progeria syndrome and hypertrophic cardiomyopathy.119,120 Thus, the therapeutic value of PTL nanoparticles for these diseases deserves further study.

The authors acknowledge funding from the Young Scientists Fund of National Science Foundation of China (No. 81803587), the Project of Science and Technology of Yunnan Province (2019FD054), PhD Start-up Fund for Tao An from Qilu University of Technology (No. 81110573), and Industry-University Cooperation Collaborative Education Project of Ministry of Education (202102403007).

The authors declare that they have no conflicts of interest in this work.

1. Vgh K, Alberti , Riethmller E, Tth A, Bni S, Kry . Supercritical fluid extraction and convergence chromatographic determination of parthenolide in Tanacetum parthenium L.: experimental design, modeling and optimization. J Supercrit Fluids. 2014;95:8491. doi:10.1016/j.supflu.2014.07.029

2. Alam P, Siddiqui NA, Rehman MT, et al. Box-Behnken Design (BBD)-Based optimization of microwave-assisted extraction of parthenolide from the stems of tarconanthus camphoratus and cytotoxic analysis. Molecules. 2021;26(7):1876. doi:10.3390/molecules26071876

3. Ghantous A, Sinjab A, Herceg Z, Darwiche N. Parthenolide: from plant shoots to cancer roots. Drug Discov Today. 2013;18(1718):894905. doi:10.1016/j.drudis.2013.05.005

4. Freund RRA, Gobrecht P, Fischer D, Arndt HD. Advances in chemistry and bioactivity of parthenolide. Nat Prod Rep. 2020;37(4):541565. doi:10.1039/C9NP00049F

5. Siveen KS, Uddin S, Mohammad RM. Targeting acute myeloid leukemia stem cell signaling by natural products. Mol Cancer. 2017;16(1):13. doi:10.1186/s12943-016-0571-x

6. Araujo TG, Vecchi L, Lima P, et al. Parthenolide and its analogues: a new potential strategy for the treatment of triple-negative breast tumors. Curr Med Chem. 2020;27(39):66286642. doi:10.2174/0929867326666190816230121

7. Dandawate PR, Subramaniam D, Jensen RA, Anant S. Targeting cancer stem cells and signaling pathways by phytochemicals: novel approach for breast cancer therapy. Semin Cancer Biol. 2016;4041:192208. doi:10.1016/j.semcancer.2016.09.001

8. Ren Y, Yu J, Kinghorn AD. Development of anticancer agents from plant-derived sesquiterpene lactones. Curr Med Chem. 2016;23(23):23972420. doi:10.2174/0929867323666160510123255

9. Mathema VB, Koh YS, Thakuri BC, Sillanpaa M. Parthenolide, a sesquiterpene lactone, expresses multiple anti-cancer and anti-inflammatory activities. Inflammation. 2012;35(2):560565. doi:10.1007/s10753-011-9346-0

10. Ghantous A, Gali-Muhtasib H, Vuorela H, Saliba NA, Darwiche N. What made sesquiterpene lactones reach cancer clinical trials? Drug Discov Today. 2010;15(1516):668678. doi:10.1016/j.drudis.2010.06.002

11. Koprowska K, Czyz M. [Molecular mechanisms of parthenolides action: old drug with a new face]. Postepy Hig Med Dosw. 2010;64:100114. Norwegian.

12. Kreuger MR, Grootjans S, Biavatti MW, Vandenabeele P, DHerde K. Sesquiterpene lactones as drugs with multiple targets in cancer treatment: focus on parthenolide. Anticancer Drugs. 2012;23(9):883896. doi:10.1097/CAD.0b013e328356cad9

13. Wyrebska A, Gach K, Janecka A. Combined effect of parthenolide and various anti-cancer drugs or anticancer candidate substances on malignant cells in vitro and in vivo. Mini Rev Med Chem. 2014;14(3):222228. doi:10.2174/1389557514666140219113509

14. Sztiller-Sikorska M, Czyz M. Parthenolide as cooperating agent for anti-cancer treatment of various malignancies. Pharmaceuticals. 2020;13(8):194. doi:10.3390/ph13080194

15. Pordanjani SM, Hosseinimehr SJ. The role of NF-kB inhibitors in cell response to radiation. Curr Med Chem. 2016;23(34):39513963. doi:10.2174/0929867323666160824162718

16. Lesiak K, Koprowska K, Zalesna I, Nejc D, Duchler M, Czyz M. Parthenolide, a sesquiterpene lactone from the medical herb feverfew, shows anticancer activity against human melanoma cells in vitro. Melanoma Res. 2010;20(1):2134. doi:10.1097/CMR.0b013e328333bbe4

17. Nasim S, Crooks PA. Antileukemic activity of aminoparthenolide analogs. Bioorg Med Chem Lett. 2008;18(14):38703873. doi:10.1016/j.bmcl.2008.06.050

18. Ren Y, Kinghorn AD. Development of potential antitumor agents from the scaffolds of plant-derived terpenoid lactones. J Med Chem. 2020;63(24):1541015448. doi:10.1021/acs.jmedchem.0c01449

19. Guzman ML, Rossi RM, Neelakantan S, et al. An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood. 2007;110(13):44274435. doi:10.1182/blood-2007-05-090621

20. Wu D, Si M, Xue HY, Wong HL. Nanomedicine applications in the treatment of breast cancer: current state of the art. Int J Nanomedicine. 2017;12:58795892. doi:10.2147/IJN.S123437

21. Zhang J, Tang H, Liu Z, Chen B. Effects of major parameters of nanoparticles on their physical and chemical properties and recent application of nanodrug delivery system in targeted chemotherapy. Int J Nanomedicine. 2017;12:84838493. doi:10.2147/IJN.S148359

22. Jasinski DL, Li H, Guo P. The effect of size and shape of RNA nanoparticles on biodistribution. Mol Ther. 2018;26(3):784792. doi:10.1016/j.ymthe.2017.12.018

23. Torchilin V. Tumor delivery of macromolecular drugs based on the EPR effect. Adv Drug Deliv Rev. 2011;63(3):131135. doi:10.1016/j.addr.2010.03.011

24. Li X, Kong L, Yang Q, et al. Parthenolide inhibits ubiquitin-specific peptidase 7 (USP7), Wnt signaling, and colorectal cancer cell growth. J Biol Chem. 2020;295(11):35763589. doi:10.1074/jbc.RA119.011396

25. Zhang S, Ju X, Yang Q, et al. USP47 maintains the stemness of colorectal cancer cells and is inhibited by parthenolide. Biochem Biophys Res Commun. 2021;562:2128. doi:10.1016/j.bbrc.2021.05.017

26. Ai XY, Zhang H, Gao SY, et al. Sesquiterpene binding Gly-Leu-Ser/Lys-co-adaptation pocket to inhibit lung cancer cell epithelial-mesenchymal transition. Oncotarget. 2017;8(41):7019270203. doi:10.18632/oncotarget.19599

27. Li X, Huang R, Li M, et al. Parthenolide inhibits the growth of non-small cell lung cancer by targeting epidermal growth factor receptor. Cancer Cell Int. 2020;20(1):561. doi:10.1186/s12935-020-01658-1

28. Garcia-Pineres AJ, Castro V, Mora G, et al. Cysteine 38 in p65/NF-kappaB plays a crucial role in DNA binding inhibition by sesquiterpene lactones. J Biol Chem. 2001;276(43):3971339720. doi:10.1074/jbc.M101985200

29. Kwok BH, Koh B, Ndubuisi MI, Elofsson M, Crews CM. The anti-inflammatory natural product parthenolide from the medicinal herb Feverfew directly binds to and inhibits IkappaB kinase. Chem Biol. 2001;8(8):759766. doi:10.1016/S1074-5521(01)00049-7

30. Kong FC, Zhang JQ, Zeng C, et al. Inhibitory effects of parthenolide on the activity of NF-kappaB in multiple myeloma via targeting TRAF6. J Huazhong Univ Sci Technolog Med Sci. 2015;35(3):343349. doi:10.1007/s11596-015-1435-0

31. Kim SL, Park YR, Lee ST, Kim SW. Parthenolide suppresses hypoxia-inducible factor-1alpha signaling and hypoxia induced epithelial-mesenchymal transition in colorectal cancer. Int J Oncol. 2017;51(6):18091820. doi:10.3892/ijo.2017.4166

32. Zhu X, Yuan C, Tian C, et al. The plant sesquiterpene lactone parthenolide inhibits Wnt/beta-catenin signaling by blocking synthesis of the transcriptional regulators TCF4/LEF1. J Biol Chem. 2018;293(14):53355344. doi:10.1074/jbc.M117.819300

33. Berdan CA, Ho R, Lehtola HS, et al. Parthenolide covalently targets and inhibits focal adhesion kinase in breast cancer cells. Cell Chem Biol. 2019;26(7):10271035 e1022. doi:10.1016/j.chembiol.2019.03.016

34. Liu M, Xiao C, Sun M, Tan M, Hu L, Yu Q. Parthenolide inhibits STAT3 signaling by covalently targeting janus kinases. Molecules. 2018;23(6):1478.

35. Gopal YN, Chanchorn E, Van Dyke MW. Parthenolide promotes the ubiquitination of MDM2 and activates p53 cellular functions. Mol Cancer Ther. 2009;8(3):552562. doi:10.1158/1535-7163.MCT-08-0661

36. Tavana O, Gu W. Modulation of the p53/MDM2 interplay by HAUSP inhibitors. J Mol Cell Biol. 2017;9(1):4552. doi:10.1093/jmcb/mjw049

37. Colleran A, Collins PE, OCarroll C, et al. Deubiquitination of NF-kappaB by Ubiquitin-Specific Protease-7 promotes transcription. Proc Natl Acad Sci U S A. 2013;110(2):618623. doi:10.1073/pnas.1208446110

38. Zhang S, Lin ZN, Yang CF, Shi X, Ong CN, Shen HM. Suppressed NF-kappaB and sustained JNK activation contribute to the sensitization effect of parthenolide to TNF-alpha-induced apoptosis in human cancer cells. Carcinogenesis. 2004;25(11):21912199. doi:10.1093/carcin/bgh234

39. Nakshatri H, Rice SE, Bhat-Nakshatri P. Antitumor agent parthenolide reverses resistance of breast cancer cells to tumor necrosis factor-related apoptosis-inducing ligand through sustained activation of c-Jun N-terminal kinase. Oncogene. 2004;23(44):73307344. doi:10.1038/sj.onc.1207995

40. DAnneo A, Carlisi D, Lauricella M, et al. Parthenolide generates reactive oxygen species and autophagy in MDA-MB231 cells. A soluble parthenolide analogue inhibits tumour growth and metastasis in a xenograft model of breast cancer. Cell Death Dis. 2013;4:e891. doi:10.1038/cddis.2013.415

41. Duan D, Zhang J, Yao J, Liu Y, Fang J. Targeting thioredoxin reductase by parthenolide contributes to inducing apoptosis of hela cells. J Biol Chem. 2016;291(19):1002110031. doi:10.1074/jbc.M115.700591

42. Pei S, Minhajuddin M, Callahan KP, et al. Targeting aberrant glutathione metabolism to eradicate human acute myelogenous leukemia cells. J Biol Chem. 2013;288(47):3354233558. doi:10.1074/jbc.M113.511170

43. Yang C, Yang QO, Kong QJ, Yuan W, Ou Yang YP. Parthenolide induces reactive oxygen species-mediated autophagic cell death in human osteosarcoma cells. Cell Physiol Biochem. 2016;40(12):146154. doi:10.1159/000452532

44. Xu Y, Fang F, Miriyala S, et al. KEAP1 is a redox sensitive target that arbitrates the opposing radiosensitive effects of parthenolide in normal and cancer cells. Cancer Res. 2013;73(14):44064417. doi:10.1158/0008-5472.CAN-12-4297

45. Liu Z, Liu S, Xie Z, et al. Modulation of DNA methylation by a sesquiterpene lactone parthenolide. J Pharmacol Exp Ther. 2009;329(2):505514. doi:10.1124/jpet.108.147934

46. Gopal YN, Arora TS, Van Dyke MW. Parthenolide specifically depletes histone deacetylase 1 protein and induces cell death through ataxia telangiectasia mutated. Chem Biol. 2007;14(7):813823. doi:10.1016/j.chembiol.2007.06.007

47. Nakshatri H, Appaiah HN, Anjanappa M, et al. NF-kappaB-dependent and -independent epigenetic modulation using the novel anti-cancer agent DMAPT. Cell Death Dis. 2015;6:e1608. doi:10.1038/cddis.2014.569

48. Fonrose X, Ausseil F, Soleilhac E, et al. Parthenolide inhibits tubulin carboxypeptidase activity. Cancer Res. 2007;67(7):33713378. doi:10.1158/0008-5472.CAN-06-3732

49. Whipple RA, Vitolo MI, Boggs AE, Charpentier MS, Thompson K, Martin SS. Parthenolide and costunolide reduce microtentacles and tumor cell attachment by selectively targeting detyrosinated tubulin independent from NF-kappaB inhibition. Breast Cancer Res. 2013;15(5):R83. doi:10.1186/bcr3477

50. Carlisi D, Lauricella M, DAnneo A, et al. Parthenolide and its soluble analogues: multitasking compounds with antitumor properties. Biomedicines. 2022;10(2):514. doi:10.3390/biomedicines10020514

51. Marino S, Bishop RT, Carrasco G, Logan JG, Li B, Idris AI. Pharmacological inhibition of NFkappaB reduces prostate cancer related osteoclastogenesis in vitro and osteolysis ex vivo. Calcif Tissue Int. 2019;105(2):193204. doi:10.1007/s00223-019-00538-9

52. Ge W, Liu Z, Sun Y, et al. Design and synthesis of parthenolide-SAHA hybrids for intervention of drug-resistant acute myeloid leukemia. Bioorg Chem. 2019;87:699713. doi:10.1016/j.bioorg.2019.03.056

53. Mehri S, Mohammadi S, Nikbakht M, Sahmani M, Zahedpanah M. Osteopontin siRNA does not confer resistance to toxic effects of parthenolide in Jurkat cells. Exp Oncol. 2020;42(3):188191. doi:10.32471/exp-oncology.2312-8852.vol-42-no-3.15180

54. Ge W, Hao X, Han F, et al. Synthesis and structure-activity relationship studies of parthenolide derivatives as potential anti-triple negative breast cancer agents. Eur J Med Chem. 2019;166:445469. doi:10.1016/j.ejmech.2019.01.058

55. De Blasio A, Di Fiore R, Pratelli G, et al. A loop involving NRF2, miR-29b-1-5p and AKT, regulates cell fate of MDA-MB-231 triple-negative breast cancer cells. J Cell Physiol. 2020;235(2):629637. doi:10.1002/jcp.29062

56. Dawood M, Ooko E, Efferth T. Collateral sensitivity of parthenolide via NF-kappaB and HIF-alpha inhibition and epigenetic changes in drug-resistant cancer cell lines. Front Pharmacol. 2019;10:542. doi:10.3389/fphar.2019.00542

57. Ding Y, Li S, Ge W, et al. Design and synthesis of parthenolide and 5-fluorouracil conjugates as potential anticancer agents against drug resistant hepatocellular carcinoma. Eur J Med Chem. 2019;183:111706. doi:10.1016/j.ejmech.2019.111706

58. Sun L, Yuan W, Wen G, et al. Parthenolide inhibits human lung cancer cell growth by modulating the IGF1R/PI3K/Akt signaling pathway. Oncol Rep. 2020;44(3):11841193. doi:10.3892/or.2020.7649

59. Luo Q, Wu X, Chang W, et al. ARID1A hypermethylation disrupts transcriptional homeostasis to promote squamous cell carcinoma progression. Cancer Res. 2020;80(3):406417. doi:10.1158/0008-5472.CAN-18-2446

60. Che ST, Bie L, Li X, Qi H, Yu P, Zuo L. Parthenolide inhibits the proliferation and induces the apoptosis of human uveal melanoma cells. Int J Ophthalmol. 2019;12(10):15311538. doi:10.18240/ijo.2019.10.03

Continue reading here:
Emerging potential of PTL nanoformulations in tumor therapy | DDDT - Dove Medical Press

Podcast: Has the First 150-Year-Old Already Been Born – Leaps

One is that there are some people that are naturally resistant to heart attack and have lifelong, low levels of LDL, the cardiologist says. Second, there are some genes that can be switched off that lead to very low LDL cholesterol, and individuals with those genes switched off are resistant to heart attacks.

Kathiresan and his team formed a hypothesis in 2016 that if they could develop a medicine that mimics the natural protection that some people enjoy, then they might identify a powerful new way to treat and ultimately prevent heart attacks. They launched Verve in 2018 with the goal of creating a one-time therapy that would permanently lower LDL and eliminate heart attacks caused by high LDL.

The medication is targeted specifically for patients who have a genetic form of high cholesterol known as heterozygous familial hypercholesterolemia, or FH, caused by expression of a gene called PCSK9. Verve also plans to develop a program to silence a gene called ANGPTL3 for patients with FH and possibly those with or at risk of atherosclerotic cardiovascular disease.

FH causes cholesterol to be high from birth, reaching levels of 200 to 300 milligrams per deciliter. Suggested normal levels are around 100 to 129 mg/dl, and anything above 130 mg/dl is considered high. Patients with cardiovascular disease usually are asked to aim for under 70 mg/dl, but many still have unacceptably high LDL despite taking oral medications such as statins. They are more likely to have heart attacks in their 30s, 40s and 50s, and require lifelong LDL control.

The goal for drug treatments for high LDL, Kathiresan says, is to reduce LDL as low as possible for as long as possible. Physicians and researchers also know that a sizeable portion of these patients eventually start to lose their commitment to taking their statins and other LDL-controlling medications regularly.

If you ask 100 patients one year after their heart attack what fraction are still taking their cholesterol-lowering medications, its less than half, says Kathiresan. So imagine a future where somebody gets a one-time treatment at the time of their heart attack or before as a preventive measure. Its right in front of us, and its something that Verve is looking to do.

In late 2020, Verve completed primate testing with monkeys that had genetically high cholesterol, using a one-time intravenous injection of VERVE-101. It reduced the monkeys LDL by 60 percent and, 18 months later, remains at that level. Kathiresan expects the LDL to stay low for the rest of their lives.

Verves gene editing medication is packaged in a lipid nanoparticle to serve as the delivery mechanism into the liver when infused intravenously. The drug is absorbed and makes its way into the nucleus of the liver cells.

Verves program targeting PCSK9 uses precise, single base, pair base editing, Kathiresan says, meaning it doesn't cut DNA like CRISPR gene editing systems do. Instead, it changes one base, or letter, in the genome to a different one without affecting the letters around it. Comparing it to a pencil and eraser, he explains that the medication erases out a letter A and makes it a letter G in the A, C, G and T code in DNA.

By making that simple change from A to G, the medication switches off the PCSK9 gene, automatically lowering LDL cholesterol.

Once the DNA change is made, all the cells in the liver will have that single A to G change made, Kathiresan says. Then the liver cells divide and give rise to future liver cells, but every time the cell divides that change, the new G is carried forward.

Additionally, Verve is pursuing its second gene editing program to eliminate ANGPTL3, a gene that raises both LDL and blood triglycerides. In 2010, Kathiresan's research team learned that people who had that gene completely switched off had LDL and triglyceride levels of about 20 and were very healthy with no heart attacks. The goal of Verves medication will be to switch off that gene, too, as an option for additional LDL or triglyceride lowering.

Success with our first drug, VERVE-101, will give us more confidence to move forward with our second drug, Kathiresan says. And it opens up this general idea of making [genomic] spelling changes in the liver to treat other diseases.

The approach is less ethically concerning than other gene editing technologies because it applies somatic editing that affects only the individual patient, whereas germline editing in the patients sperm or egg, or in an embryo, gets passed on to children. Additionally, gene editing therapies receive the same comprehensive amount of testing for side effects as any other medicine.

We need to continue to advance our approach and tools to make sure that we have the absolute maximum ability to detect off-target effects, says Euan Ashley, professor of medicine and genetics at Stanford University and founding director of its Center for Inherited Cardiovascular Disease. Ashley and his colleagues at Stanfords Clinical Genomics Program and beyond are increasingly excited about the promise of gene editing.

We can offer precision diagnostics, so increasingly were able to define the disease at a much deeper level using molecular tools and sequencing, he continues. We also have this immense power of reading the genome, but were really on the verge of taking advantage of the power that we now have to potentially correct some of the variants that we find on a genome that contribute to disease.

He adds that while the gene editing medicines in development to correct genomes are ahead of the delivery mechanisms needed to get them into the body, particularly the heart and brain, hes optimistic that those arent too far behind.

It will probably take a few more years before those next generation tools start to get into clinical trials, says Ashley, whose book, The Genome Odyssey, was published last year. The medications might be the sexier part of the research, but if you cant get it into the right place at the right time in the right dose and not get it to the places you dont want it to go, then that tool is not of much use.

Medical experts consider knocking out the PCSK9 gene in patients with the fairly common genetic disorder of familial hypercholesterolemia roughly one in 250 people a potentially safe approach to gene editing and an effective means of significantly lowering their LDL cholesterol.

Nurse Erin McGlennon has an Implantable Cardioverter Defibrillator and takes medications, but she is also hopeful that a gene editing medication will be developed in the near future.

Erin McGlennon

Mary McGowan, MD, chief medical officer for The Family Heart Foundation in Pasadena, CA, sees the tremendous potential for VERVE-101 and believes patients should be encouraged by the fact that this kind of research is occurring and how much Verve has accomplished in a relatively short time. However, she offers one caveat, since even a 60 percent reduction in LDL wont completely eliminate the need to reduce the remaining amount of LDL.

This technology is very exciting, she said, but we want to stress to our patients with familial hypercholesterolemia that we know from our published research that most people require several therapies to get their LDL down., whether that be in primary prevention less than 100 mg/dl or secondary prevention less than 70 mg/dl, So Verves medication would be an add-on therapy for most patients.

Dr. Kathiresan concurs: We expect our medicine to lower LDL cholesterol by about 60 percent and that our patients will be on background oral medications, including statins that lower LDL cholesterol.

Several leading research centers are investigating gene editing treatments for other types of cardiovascular diseases. Elizabeth McNally, Elizabeth Ward Professor and Director at the Center for Genetic Medicine at Northwestern Universitys Feinberg School of Medicine, pursues advanced genetic correction in neuromuscular diseases such as Duchenne muscular dystrophy and spinal muscular atrophy. A cardiologist, she and her colleagues know these diseases frequently have cardiac complications.

Even though the field is driven by neuromuscular specialists, its the first therapies in patients with neuromuscular diseases that are also expected to make genetic corrections in the heart, she says. Its almost like an afterthought that were potentially fixing the heart, too.

Another limitation McGowan sees is that too many healthcare providers are not yet familiar with how to test patients to determine whether or not they carry genetic mutations that need to be corrected. We need to get more genetic testing done, she says. For example, thats the case with hypertrophic cardiomyopathy, where a lot of the people who probably carry that diagnosis and have never been genetically identified at a time when genetic testing has never been easier.

One patient who has been diagnosed with hypertrophic cardiomyopathy also happens to be a nurse working in research at Genentech Pharmaceutical, now a member of the Roche Group, in South San Francisco. To treat the disease, Erin McGlennon, RN, has an Implantable Cardioverter Defibrillator and takes medications, but she is also hopeful that a gene editing medication will be developed in the near future.

With my condition, the septum muscles are just growing thicker, so Im on medicine to keep my heart from having dangerous rhythms, says McGlennon of the disease that carries a low risk of sudden cardiac death. So, the possibility of having a treatment option that can significantly improve my day-to-day functioning would be a major breakthrough.

McGlennon has some control over cardiovascular destiny through at least one currently available technology: in vitro fertilization. Shes going through it to ensure that her children won't express the gene for hypertrophic cardiomyopathy.

Go here to see the original:
Podcast: Has the First 150-Year-Old Already Been Born - Leaps

Tackling chronic disease with gene and cell therapies – The Irish Times

The repair of old, damaged, or diseased tissues using gene or cell therapies promises a future where people live longer, healthier lives and Ireland is well placed to become a manufacturing hub for products based on this technology.

Gene therapy is the technology used to correct a gene defect that is causing an inherited genetic disease. Cell therapy is the use of living cells from the patient or a donor, to repair tissue or treat an inflammatory condition or disease. These therapies can be used alone or combined for greater effect.

Weve been interested, for a very long time in the development of new treatments for patients involving cell and gene therapy, says professor of cellular therapy at NUI Galway Frank Barry a co-founder of the Regenerative Medicine Institute (Remedi) in 2004.

Over the last several decades there have been some extraordinary, transformative developments in medicine; for example, antibiotics and monoclonal antibodies and these have had a dramatic impact on how diseases were treated, says Barry. Many people believe that cell and gene therapy represent the next transformative innovation that will change medicine.

There are many examples of outstanding success stories, where diseases which were previously untreatable are now actually being treated and were very anxious to continue to play a role in this, says Barry.

The combination of cell and gene therapy has been successful in treating cancers that were thought incurable. For example, stem cells have been taken from the blood of patients with specific cancers, genetically modified so they target a particular cancer, and are delivered back into the patients blood.

Gene therapy has had a troubled history with some adverse outcomes reported from early clinical trials two decades ago. Most notably, and tragically, was the case of 18-year-old Jesse Gelsinger, who died in 1999 during a University of Pennsylvania run gene therapy trial. Jesse suffered from a genetic disease affecting his liver which meant that he was unable to metabolise ammonia.

The learnings from that have proven to be very helpful not to diminish the impact of his death on his family and the tragedy of that, says Prof Tim OBrien, head of medicine at NUI Galway, an Irish pioneer of this field.

In Ireland, the origins of cell and gene therapy research go back to 2004, when Remedi was set up with funding from Science Foundation Ireland (SFI). Then in 2014, the Centre for Cell Manufacturing in Ireland (CCMI) was established. Barry and OBrien have been the key figures driving the process.

From the beginning, the dream of Barry and OBrien was to convert promising gene and cell therapy research into new therapies that could then be tested in clinical trials. Galway was a good place to do it, as it was known as a leading hub for medical device research and manufacturing, and it had the laboratories, hospital tissue facilities and clinical trial expertise that would be required.

Almost two decades down the road, the next step, they say, requires putting in place a national plan for developing a cell and gene therapy industry across the island similar to what has been achieved for medical devices and other high areas dependent on advanced technology, like ICT and pharmaceuticals. The UK offers a model of what can be achieved as it benefits from a decision by government to heavily invest in gene and cell therapy 15 years ago through an independent body it established called the Cell and Gene Therapy Catapult.

The opportunity for gene and cell therapy to grow here in coming years helped attract Dr Meadhbh Brennan, a post-doctoral researcher at Harvard University, back to Ireland. She also worked at the National Institute of Health and Inserm in France before returning to NUIG to set up her own research group.

In France, Brennan had worked on a clinical trial using stem cells to treat bone defects, while in the US her research focused on factors secreted by stem cells which could be used as a therapeutic. While in the US, she was awarded funding from SFI and that provided impetus for her move home, to take up a position at NUIG working at the interface between engineering and medicine.

She has a European Research Council starting grant award to investigate ways of regenerating bone defects, building on her work in this area. There are more than one million bone grafting procedures performed annually in Europe, and after blood, bone is the most transplanted tissue. There are issues with these procedures, however, as bone tissue is limited in quantity and quality and there is often pain at the surgical site for patients.

Brennan and her team are seeking alternatives to bone grafting through the use of byproducts from the manufacturing of stem cells called extracellular vesicles (EVs). These EVs are tiny biological packages that each contain a therapeutic cargo that has been shown to be capable of enhancing healing processes in tissues by delivering healing messages from cell to cell.

Up to now, EVs have been disposed of as waste products from commercial stem cell manufacturing. We want to divert these discarded products and harness their therapeutic potential, Brennan says, This will make the whole stem cell manufacturing process more efficient and sustainable.

Remedi scientists have experience running patient cell therapy trials, with a trial to treat arthritis of the knee using patients own cells having finished and its results set to be reported during 2022. We dont have the formal results yet, but every piece of information that weve seen about this kind of effort suggests that there is a positive benefit associated with delivering cells to these arthritic joints, Barry says. The next step would be to conduct a larger, well controlled, multinational trial of the therapy, which could be led in Ireland.

There is a huge need for new therapies to treat bone defects, given that about 10 per cent of all bone fractures wont heal if left alone, while bone infection and surgery can leave big voids in bone that need to be healed. This is where new approaches based on EVs can come in, says Brennan. These tiny particles hold huge promise for regenerating not only bone tissue, but also older tissues and organs, and have healing potential in other diseases too.

A key challenge to sort out with EV-based therapies is to find a way to safely transport them from the stem cell manufacturing facility to the clinic. Brennan and her team are investigating ways to allow the vesicles to be stored for longer durations at room temperature. The ultimate goal is to develop novel EV treatments that are inexpensive and available off the shelf when a patient has an injured or damaged tissue or needs an anti-inflammatory treatment.

The whole idea about these technologies is that they are regenerative, stimulate repair or correct defects which are chronic, Barry notes. If they work then you are saving years and years of care associated with chronic illness. The economics of this make an awful lot of sense, and the investment that is needed is very much worth it in terms of the long term.

This is a huge new industry which is exploding worldwide and will require advanced manufacturing capacity in all corners of the world, Barry points out. There is an opportunity for Ireland to become a major centre of this, and we have the people, expertise, and infrastructure to allow the industry to develop here. We need to move with lightning speed to capture the opportunity.

The idea of Ireland becoming a global hub for cell and gene therapy and manufacturing is something we should talk about seriously, he adds.

Read more here:
Tackling chronic disease with gene and cell therapies - The Irish Times

Gamida Cell Announces Results of New Health Economic and Outcome Study Reporting Improved Health Equity – BioSpace

BOSTON--(BUSINESS WIRE)-- Gamida Cell Ltd.. (Nasdaq: GMDA), the leader in the development of NAM-enabled cell therapies for patients with solid and hematological cancers and other serious diseases, today announced the results of a new study demonstrating the potential impact of access to omidubicel on health disparities in allogeneic hematopoietic stem cell transplant in a poster presentation at the 2022 Transplantation & Cellular Therapy Meetings of ASTCT and CIBMTR Tandem Meetings (TCT), being held in Salt Lake City, UT, April 23-26, 2022.

The study, titled Projected Impact of Omidubicel on Racial and Ethnic Disparities in Allogeneic Hematopoietic Cell Transplant Access and Outcomes for Patients with Hematologic Malignancies in the US, leveraged a decision-tree model to project allo-HCT access and clinical outcomes in a hypothetical population of 10,000 allo-HCTeligible patients in the U.S. with hematologic malignancies without an available match-related donor. The study concluded that broad use of omidubicel will extend access for allo-HCT-eligible patients, decrease time to transplant and improve clinical outcomes, notably among racial and ethnic groups with worse status quo outcomes. Projected increases in one-year overall survival ranged (with 20% omidubicel use among allo-HCTeligible patients) from 2.5% for whites patients to 6.3% for Black patients. The study also concluded that higher levels of modeled omidubicel uptake were associated with greater improvements in clinical outcomes and greater reductions in racial disparities.

Previous studies indicate that non-white patients have a lower likelihood of finding an appropriate match in the U.S. public donor registries, with Black patients have a 16-20% chance of finding an appropriate match. Given that an allogeneic stem cell transplant is intended as a curative option, if patients cannot find an appropriate match they will not have access to allogeneic stem cell transplant, a potentially curative treatment. The Phase 3 study of omidubicel demonstrated the ability of the therapy to be used as a donor source for racially and ethnically diverse patients with 40% of patients enrolled in the study being non-white.

Today, minority groups comprise only about 30% of all allogeneic hematopoietic stem cell transplant transplants, indicating that lack of access to a matched donor is a significant barrier to treatment in the current landscape, said Julian Adams, Ph.D., Chief Executive Officer of Gamida Cell. This study is encouraging in that it projects that broad access to omidubicel has the potential to open up allo-HSCT as an effective treatment for more patients and address the barriers that have contributed to this alarming health disparity. These data are particularly encouraging as we continue to advance our rolling BLA submission to the FDA and move closer to bringing the therapy to more patients in need.

Gamida Cell initiated a rolling Biologics License Application (BLA) submission for omidubicel in the first quarter of 2022 and is on-track to complete submission of all modules of the BLA in the second quarter of 2022.

In addition to this poster, two oral presentations and four additional poster presentations on omidubicel and a poster presentation on GDA-201, the companys leading NK cell therapy program, will be shared during the conference. All poster presentations will be publicly available at http://www.ASTCT.org. Details below:

About Omidubicel

Omidubicel is an advanced cell therapy under development as a potential life-saving allogeneic hematopoietic stem cell transplant for patients with hematologic malignancies (blood cancers), for which it has been granted Breakthrough Status and orphan drug designation by the FDA. Omidubicel is also being evaluated in a Phase 1/2 clinical study in patients with severe aplastic anemia (NCT03173937). For more information on clinical trials of omidubicel, please visit the Gamida Cell website.

Omidubicel is an investigational therapy, and its safety and efficacy have not been established by the FDA or any other health authority.

About NAM Technology

Our NAM-enabling technology is designed to enhance the number and functionality of targeted cells, enabling us to pursue a curative approach that moves beyond what is possible with existing therapies. Leveraging the unique properties of NAM (nicotinamide), we are able to enhance, expand and metabolically modulate multiple cell types including stem cells and natural killer cells with appropriate growth factors to maintain the cells active phenotype and enhance potency. This allows us to administer a therapeutic dose of cells that may help cancer patients live longer better lives.

About Gamida Cell

Gamida Cell is pioneering a proprietary NAM-enabled immunotherapy pipeline of diverse potentially curative cell therapies for patients with solid tumor and blood cancers and other serious blood diseases. We apply a proprietary platform leveraging the properties of NAM to allogeneic cell sources including umbilical cord blood-derived cells and NK cells to create therapies with potential to redefine standards of care. These include omidubicel, an investigational product with potential as a life-saving alternative for patients in need of transplant, and a line of modified and unmodified NAM-enabled NK cells targeted at solid tumor and hematological malignancies. For additional information on Gamida Cell, please visit http://www.gamida-cell.com or follow Gamida Cell on LinkedIn, Twitter, Facebook or Instagram at @GamidaCellTx.

Cautionary Note Regarding Forward Looking Statements

This press release contains forward-looking statements as that term is defined in the Private Securities Litigation Reform Act of 1995, including with respect to timing of initiation and progress of, and data reported from, the clinical trials of Gamida Cells product candidates (including GDA-201), anticipated regulatory filings and the potentially life-saving or curative therapeutic and commercial potential of omidubicel. Any statement describing Gamida Cells goals, expectations, financial or other projections, intentions or beliefs is a forward-looking statement and should be considered an at-risk statement. Such statements are subject to a number of risks, uncertainties and assumptions, including those related to the impact that the COVID-19 pandemic could have on our business, and including the scope, progress and expansion of Gamida Cells clinical trials and ramifications for the cost thereof; clinical, scientific, regulatory and technical developments; and those inherent in the process of developing and commercializing product candidates that are safe and effective for use as human therapeutics, and in the endeavor of building a business around such product candidates. In light of these risks and uncertainties, and other risks and uncertainties that are described in the Risk Factors section and other sections of Gamida Cells Annual Report on Form 10-K, filed with the Securities and Exchange Commission (SEC) on March 24, 2022, as amended, and other filings that Gamida Cell makes with the SEC from time to time (which are available at http://www.sec.gov), the events and circumstances discussed in such forward-looking statements may not occur, and Gamida Cells actual results could differ materially and adversely from those anticipated or implied thereby. Although Gamida Cells forward-looking statements reflect the good faith judgment of its management, these statements are based only on facts and factors currently known by Gamida Cell. As a result, you are cautioned not to rely on these forward-looking statements.

View source version on businesswire.com: https://www.businesswire.com/news/home/20220425005348/en/

Here is the original post:
Gamida Cell Announces Results of New Health Economic and Outcome Study Reporting Improved Health Equity - BioSpace

We have found an antibody against cancer stem cells. Its less exciting than it seems – InTallaght

This week, a piece of news about cancer has found its way into the dense web of current political, economic and military information: an international consortium led by the Barcelona Institute for Biomedical Research (IRB) has just completed the pre-clinical studies that place the MCLA-158 antibody at the head of the race to find the first drug to target cancer stem cells from solid tumors.

Cancer stem cells? Until recently, we believed that cancer was essentially a homogeneous mass of rapidly proliferating cells. For this reason, the therapies that were emerging focused on that: on eliminating those highly proliferative cells. In recent decades, we have learned that there is enormous diversity in cancer cells. Both in proliferation and differentiation.

Whats more, in recent years, we discovered cancer stem cells. A small subset of cells that, as in the case of normal stem cells, have the capacity to renew and generate the variety of proliferating and differentiated cells that make up the bulk of the tumor. The problem is that they are not affected by the therapies we used. To put it metaphorically, we were attacking the soldiers, but the factory was still running.

How to differentiate a normal stem cell from a cancerous one? Every time we discovered this problem, researchers have focused on finding mechanisms that allow us to attack some cells without compromising the rest. In this case, MCLA-158 is a bispecific antibody that recognizes two characteristic proteins of cancer stem cells (EGFR and LGR5). The idea of the team led by Eduard Batlle is that, precisely for this reason, it should not interfere with the functioning of the bodys healthy stem cells, which are essential for the proper functioning of tissues.

Still far from the clinic. This is a very important discovery and there are indications that the data is strong, but we need to temper our enthusiasm. These are preclinical data. We have known for a long time that only 5% of all drugs that have demonstrated their effectiveness in preclinical phases reach the market. As I say, this is not an argument against this antibody; it is rather an argument against overly sensational narratives.

welcome organoids. However, there is something for which this work is interesting beyond what happens in the future: the use of organoids. The researchers built a biobank that features organoids derived from colon cancer patients, organoids from colon cancer metastases in the liver, and organoids from normal, noncancerous tissue.

Organoids explained to us a few years ago Benjamin Freedman, professor of medicine at the University of Washington and an expert in kidney organoids are collections of cells on a support, like a plate, that resemble a tissue or organ of the body . This means that, by incorporating them in the earliest phases of drug generation, he helps identify those that are effective for most patients or even for tumors that carry a particular mutation. It allows us to go faster.

At least that was what we believed. Now, with the good results of this study and the development of a clear methodology for the use of organoids, we can confirm it. In this way, the work of Batlle and his team opens the door to better, faster and more efficient cancer science. Even if the antibody ultimately failed to reach hospitals, its contribution would have been enormous.

Read the original:
We have found an antibody against cancer stem cells. Its less exciting than it seems - InTallaght

I would not be here were it not for the blood: Duval residents describe impact of blood donation – Yahoo News

Community members are speaking out about the role blood donation has played in their personal lives.

This comes as Action News Jax and our Family Focus partners have teamed up for our annual Spring Into Action Blood Drive.

Jacksonville resident Penelope McGowan told Action News Jax reporter Kennedy Dendy that having the opportunity to give blood is an honor. My father needed a life-saving procedure, so it became more important to me to start giving blood, McGowan said.

She then became a regular giver, knowing the impact donation truly has.

That allowed him the time to spend time with his grandchildren, McGowan said. He walked his granddaughter down the aisle and got to see some of his great-grandchildren.

McGowan said blood donation made that moment possible.

Now that hes passed away, I want to give that gift of time to other families, McGowan said. So, its so important to me to give blood.

RELATED: OneBlood, Action News Jax team up for the Spring into Action Blood Drive

Action News Jax also spoke with John Dean, who is a patient at the Mayo Clinic. Hes from South Carolina but has been living in Jacksonville since January.

I got the bone marrow transplant, which is basically a stem cell infusion on January 17th, Dean said. I have been dealing with myelodysplastic syndrome.

Dean said its a form of bone marrow cancer hes been battling since 2017.

During that time, I had become increasingly dependent upon blood because the syndrome destroys my bodys ability to make red blood cells, Dean said. So when the blood numbers drop, I get very very sick.

He said the transplant was designed to cut down on his need to get the blood, but that hasnt happened yet.

Ive been more blood dependent since January than I had been before I came down here, Dean said.

Dean spoke with me just moments after he received a blood transfusion at the hospital -- but he wanted one message out there.

Youre transmitting a miracle, Dean said. Im a living example of that. I would not be here were it not for the blood.

Story continues

OneBlood said to donate youll need an ID, and you must be 16 years and older.

Randy Varner donated double red blood cells at Tuesdays drive.

My wife has had to have two heart valves replaced, so shes had to have blood before at the hospital -- so I try to help out when I can, Varner said.

Varner shared that if youre able to -- you should give.

Theres nothing to it, Varner said. You go in there. You answer a few questions. You lay down. You can take a little nap if you have to.

Nicole Payne is the Senior Program and Membership Director with the Brooks Family YMCA, one of the many sites for the drive.

Theres always a lack of blood available for people that come into any traumatic situation, Payne said. We want to make sure that we can hopefully combine some of the best parts of Jacksonville -- and thats through OneBlood being here to help people have access to donate.

The Spring into Action Blood Drive kicked off Tuesday and runs through Friday.

When you donate you will receive a free t-shirt, a $20 e-gift card, and an additional gift depending on the location where you donate.

CLICK HERE to find out when and where you can donate.

STAY UPDATED: Download the Action News Jax app for live updates on breaking stories

Download WJAX Apps

More:
I would not be here were it not for the blood: Duval residents describe impact of blood donation - Yahoo News

Quell Therapeutics and Cellistic enter a strategic collaboration to develop an iPSC-derived allogeneic T-regulatory (Treg) cell therapy platform – PR…

- Collaboration combines Quell's pioneering autologous multi-modular Treg cell therapy platform and Cellistic's expertise in iPSC cell therapy platform development and scale-up

- Aims to accelerate the development of a next-generation allogeneic Treg platform that could open significant opportunities for Quell's creation of off-the-shelf Treg cell therapies targeting a wide range of diseases driven by immune dysregulation

- First announced collaboration for Cellistic, Ncardia's recently formed cell therapy process development and manufacturing services business

LONDON and GOSSELIES, Belgium, April 27, 2022 /PRNewswire/ --Quell Therapeutics Ltd ("Quell"), a leader in developing engineered T-regulatory (Treg) cell therapies for serious medical conditions driven by the immune system, and Cellistic,the iPSC-focused cell therapy process development & manufacturing partner recently launched by Ncardia to make large-scale allogeneic cell therapy production a reality,announce they have entered into a strategic collaboration for the co-development of an iPSC-derived Treg cell therapy platform. The goal of the partnership is to facilitate the future expansion of Quell's autologous Treg cell therapy pipeline by adding off-the-shelf, allogeneic Treg cell therapy products, leveraging Cellistic's expertise in differentiation and scale-up of iPSC processes for allogeneic cell therapy applications.

Iain McGill, Chief Executive Officer, Quell Therapeutics,said: "Quell has made significant progress advancing the first candidate from our autologous multi-modular Treg cell therapy platform into the clinic, with the initiation of our LIBERATE study of QEL-001 to prevent liver transplant rejection. We believe there is significant opportunity to transform outcomes for patients with QEL-001 and other autologous Treg cell therapy products in our pipeline. Our collaboration with Cellistic is a key building block in our investment towards a future, next-generation allogeneic Treg cell platform, which could significantly expand our opportunities to develop novel off-the-shelf treatments across a wide range of diseases driven by immune dysregulation. We highly respect the expertise and experience of Ncardia and the Cellistic team, and its track record in developing rapidly scalable iPSC cell therapy processes."

Stefan Braam, Chief Executive Officer, Cellistic,said: "Our partnership with Quell is emblematic of why we started Cellistic to bring together our focus and expertise in the development and implementation of iPSC cell therapy platforms with companies like Quell that have an equal depth of expertise in therapeutic development and share our vision for the future of cell therapy. We are excited to collaborate with the Quell team, both to develop the platforms, and to support Quell's long-term supply needs as they deliver impactful therapeutics to patients."

Under the terms of the agreement, Quell and Cellistic will collaborate in joint research to develop a process for differentiating iPSCs into highly functional Treg cell therapy products. Quell will contribute its Treg expertise and engineering technologies, as well as characterizing resulting Treg cells, while Cellistic will be responsible for the iPSC process science and development activities.

Based on a successful research phase, the collaboration will enter a product development phase with Quell having exclusive rights under the co-developed iPSC-Treg process for the development of multiple allogeneic iPSC-Treg cell therapeutics, and Cellistic as the exclusive CDMO partner for Quell's iPSC-Treg product pipeline, leveraging Cellistic's ongoing investment in downstream GMP capabilities.

Tracey Lodie, Chief Scientific Officer, Quell Therapeutics,added: "We have learnt in cell therapy development that the continuity from the R&D phase into the manufacturing phase is a critical success factor in achieving robust, scalable cell product processes. Cellistic emerged as a best-in-class and complementary partner to enable our path to an iPSC Treg cell therapy platform, and its ongoing investment in GMP capabilities provides the potential for a long-term partnership to accelerate the future development of allogeneic Treg cell therapies for patients."

About Quell Therapeutics

Quell Therapeutics is the world leader in developing engineered T-regulatory (Treg) cell therapies that aim to harness, direct and optimize their immune suppressive properties to address serious medical conditions driven by the immune system.

The Company is leveraging its pioneering phenotype lock technology, unique multi-modular platform and integrated manufacturing capabilities to design and develop a pipeline of highly engineered Treg cell therapies with greater potential for persistence, potency and stability than earlier generations of Treg cell therapy approaches.

Quell's lead candidate QEL-001 is being developed to induce operational tolerance following liver transplantation, with the potential to protect the post-transplant liver without the need for chronic immunosuppressive medications. Quell is also advancing additional programs in neuroinflammatory and autoimmune diseases. http://www.quell-tx.com.

About Cellistic

Launched in April 2022, Cellistic specializes in process development and manufacture of cell therapies based on human induced pluripotent stem cell (iPSC) technology. Its focus and expertise in iPSC reprogramming, differentiation, and expansion protocol development positions the business to be the partner of choice for innovative cell therapy developers to commercialize novel advanced therapies. Leveraging more than a decade of Ncardia's scientific and technical knowledge and experience, Cellistic possesses unique capabilities for the design and optimization of proprietary manufacturing platforms for iPSC-based cells that deliver quality products at scale. For more information, visit http://www.cellistic.com.

About Ncardia

Ncardia is a leader in contract research, development and manufacture of iPSC-based solutions for early and preclinical drug discovery. Its goal is to enable pharmaceutical and therapeutics companies to make more confident decisions in discovery and development by integrating iPSC technologies into their screening processes. Ncardia's capabilities include disease modeling, manufacturing, assay development and high-throughput screening especially for cardiac and neurodegenerative diseases. Ncardia was founded in 2011 and is majority-owned by KINICITI a private equity-backed advanced therapies platform. For more information, visit http://www.ncardia.com

Contacts for Quell TherapeuticsLuke Henry, Chief Business Officer Quell Therapeutics[emailprotected]

Media: Mark Swallow, Sandi Greenwood, Eleanor Perkin MEDiSTRAVA Consulting+44 203 928 6900 [emailprotected]

Investors: Christina Tartaglia Stern Investor Relations, Inc.+1 212 362 1200 [emailprotected]

Contacts for Ncardia/CellisticAndy Holt, Chief Commercial Officer Cellistic[emailprotected]

SOURCE Quell Therapeutics and Cellistic

See original here:
Quell Therapeutics and Cellistic enter a strategic collaboration to develop an iPSC-derived allogeneic T-regulatory (Treg) cell therapy platform - PR...

Global Cell Therapy Market to be Driven by Rapidly Growing Research and Development (R&D) Efforts and Advantageous Government Initiatives During the…

The new report by Expert Market Research titled, Global Cell Therapy Market Report and Forecast 2021-2026, gives an in-depth analysis of the global cell therapy market, assessing the market based on its segments like applications, manufacturing process and major regions. The report tracks the latest trends in the industry and studies their impact on the overall market. It also assesses the market dynamics, covering the key demand and price indicators, along with analyzing the market based on the SWOT and Porters Five Forces models.

Get a Free Sample Report with Table of Contents: https://www.expertmarketresearch.com/reports/cell-therapy-market/requestsample

The key highlights of the report include:

Market Overview (2016-2026)

The industry is being propelled forth by increasing number of clinical trials for mitochondrial medicines. As a result, global investment in research and clinical application has increased significantly. The presence of government and commercial funding agencies that are constantly awarding grants to assist clinical trials at various stages can be attributed to the expanding number of ongoing clinical trials. Additionally, market growth has been aided by the continuous introduction of new technologies that have facilitated the creation of novel medicines. The introduction of proprietary cell lines is often recognized as the most successful method for generating a diversified variety of alternatives from a single product. During the forecast period, this type of continual activity is likely to propel the markets growth.

Industry Definition and Major Segments

Cell therapy (also known as organic transplantation or cytotherapy) is a type of therapy in which viable cells are implanted, grafted, or infused into a patient to achieve a therapeutic effect, such as by transplanting T-cells capable of fighting cancer cells via cell-mediated immunity during immunotherapy or by grafting stem cells to regenerate diseased tissues.

Read Full Report with Table of Contents: https://www.expertmarketresearch.com/reports/cell-therapy-market

By type, the market is divided into:

By therapies, the market is divided into:

By region, the industry is categorized into:

Market Trends

The medical sectors growing research and development (R&D) efforts are driving the market. The pandemic of COVID-19 is projected to have hastenedpharmacological and biotechnological research efforts, including cell therapy. Because the lethal virus severely destroys the infected patients lungs and immune system, numerous cell treatments are increasingly being investigated as a viable cure for the unique disease, boosting market growth. Another breakthrough for the cell therapy market is expected to be the development of an effective treatment for solid tumors. In the forecast period, the potential for cell therapy to cure significant diseases such as anemia, cancer, and Alzheimers disease, among others, is expected to boost the cell therapy business. The research-use category led the cell therapy market, accounting for the majority of the market share. Replacement, repair, restoration, and regeneration of damaged tissues, cells, and organs are all possibilities with cell-based therapies. As an alternative to standard treatment procedures, researchers are investing heavily in the creation of effective and safe remedies. The market for cell treatment is being driven by the growth of cell banking and companies shift toward the development of allogenic therapeutic products.

Due to the high prevalence of genetic illnesses, North America dominates the global cell and gene therapy market. The regions large share can be linked to the presence of a large number of centers and institutes working in stem cell treatment research and development. Over the projected period, the market for cell therapy in Asia and the Pacific is expected to develop at a robust pace. The growing investment by developers and consumers in self-education for advanced medicinal therapies is expected to improve revenue generated by the regions emerging economies. In Asian countries, there are a number of stem cell consortiums whose goal is to ensure that R&D projects are well-coordinated and targeted. Furthermore, because of the flexibility in the legal framework, many patients from western countries have been spotted migrating to these Asian countries for their treatment. Factors such as advantageous government initiatives and an increase in the number of research collaborations among top market players are expected to drive the growth of the global market.

Latest News on Cell Therapy Market: https://www.expertmarketresearch.com/pressrelease/global-cell-therapy-market

Key Market Players

The major players in the market are Pfizer Inc., F. Hoffmann-LA Roche Ltd, Sanofi SA, Bristol-Myers Squibb Company, AbbVie Inc., Novartis AG, GlaxoSmithKline PLC, Amgen Inc., Johnson & Johnson, and Takeda Pharmaceutical Co. Ltd, among Others. The report covers the market shares, capacities, expansions, investments and mergers and acquisitions, among other latest developments of these market players.

About Us:

Expert Market Research is a leading business intelligence firm, providing custom and syndicated market reports along with consultancy services for our clients. We serve a wide client base ranging from Fortune 1000 companies to small and medium enterprises. Our reports cover over 100 industries across established and emerging markets researched by our skilled analysts who track the latest economic, demographic, trade and market data globally.

At Expert Market Research, we tailor our approach according to our clients needs and preferences, providing them with valuable, actionable and up-to-date insights into the market, thus, helping them realize their optimum growth potential. We offer market intelligence across a range of industry verticals which include Pharmaceuticals, Food and Beverage, Technology, Retail, Chemical and Materials, Energy and Mining, Packaging and Agriculture.

Media Contact

Company Name: EMR Inc. Contact Person:-Ian Bell, Business Consultant Email:sales@expertmarketresearch.com Toll Free Number: US +1-415-325-5166 | UK +44-702-402-5790 Address: 30 North Gould Street, Sheridan, WY 82801, USA City: Sheridan State: Wyoming Country: United States Website:https://www.expertmarketresearch.com

Read More Reports:

Global Thrombin Market: https://www.expertmarketresearch.com/reports/thrombin-market

Global Polymeric Biomaterials Market: https://www.expertmarketresearch.com/reports/polymeric-biomaterials-market

Global PD-1 and PD-L1 Inhibitors Market: https://www.expertmarketresearch.com/reports/pd-1-and-pd-l1-inhibitors-market

Global Biosurgery Market: https://www.expertmarketresearch.com/reports/biosurgery-market

Global Albumin Market: https://www.expertmarketresearch.com/reports/albumin-market

Explore more onProcurement Intelligenceservices of EMR Inc.

**We at Expert Market Research always thrive to give you the latest information. The numbers in the article are only indicative and may be different from the actual report.

Go here to read the rest:
Global Cell Therapy Market to be Driven by Rapidly Growing Research and Development (R&D) Efforts and Advantageous Government Initiatives During the...

3D Cell Culture Market is Set to Experience a Significant Growth of 9.8% CAGR from 2022 To 2031 – Comprehensive Research Report By FMI – BioSpace

DUBAI, United Arab Emirates, The 3D Cell Culture Market revenues surpassed US$ 1.15 Billion in 2022, as per a new FMI study. The market is estimated to grow at 7.8% y-o-y in 2019.

3D Cell Culture Market is predicted to surge ahead at a CAGR of 9.8% over the forecast period to attain a valuation of US$ 2.67 Billion by 2031

key factors responsible for the projected market growth include,

Request a report sample to gain comprehensive insights@ https://www.futuremarketinsights.com/reports/sample/rep-gb-2843

While 2D cell culture revolutionized the research efforts in stem cells, tissue engineering, and molecular biology, 3D cell culture has pushed the boundaries of traditional 2D cell culture technique with functional superiority. As the R&D efforts continue to rise in a bid to investigate the cause of different diseases and improve human health, 3D cell culture is set to remain a highly sought-after technique in the coming years, says FMI.

The FMI study finds that scaffold-based 3D cell culture techniques are highly preferred over scaffold-free 3D cell culture. Owing to the significant adoption of scaffold-based 3D cell culture, the study finds that this technique garnered over 81% of the global market revenues.

Scaffold-based 3D cell culture techniques deliver researchers with additional functional operations in terms of material natural or synthetic and different mechanical properties.

The technique uses either hydrogel-based support or polymeric hard material based support. Both types of supports find equivalent penetration in terms of application, however revenues garnered from polymeric hard material based support are higher.

According to the study, revenues of polymeric hard material based support held over half the scaffold-based 3D cell culture technique revenues in 2018 and the trend is expected to continue in the future.

For any Queries Related with the Report, Ask an Analyst @ https://www.futuremarketinsights.com/ask-question/rep-gb-2843

Application of 3D Cell Culture in Cancer Research Prominent

The FMI study estimates that 3D cell culture application in cancer research accounted for over 31% of the 3D cell culture market revenues in 2018. Cell culture is an integral part of cancer drug discovery practices. Greater strides are underway in the field to precisely characterize the diseases and develop advanced tumor cell lines using 3D cell culture techniques.

2D culture lines are considered as a standard for in vitro pre-clinical cancer treatment screening. However, more recently, the field is turning to 3D cell culture techniques to implement an ideal experimental model that mimics the human body environment to its best.

Stem cell technology is another lucrative field for 3D cell culture market. According to the FMI study, application in stem cell technology accounted for over one-fourth of the 3D cell culture market revenues in 2018. While 2D cell culture posed challenges of scalability in stem cell technology, apart from a few challenges, 3D cell culture has provided greater density and multi-fold expansion of the culture system in stem cell technology. Other fields that utilize 3D cell culture techniques are tissue regeneration, regenerative medicine, and drug discovery.

Biotechnology and Pharmaceutical Industries - Prominent End-Users

The study reveals that the biotechnology and pharmaceutical industries were accounted for over one-fifth of the global 3D cell culture market revenues in 2018. Buoyancy in the biotechnology and pharmaceutical industries can be profitable for the 3D cell culture market in the coming years and over 9.8% growth is projected in 2022 over 2031.

Academic research institutes are the second prominent end-users of 3D cell cultures wherein they accounted for over one-fourth of the global 3d cell culture revenues in 2018. Government funding in cancer research projects carried out at academic institutes and organizations has increased significantly, thereby supporting the investments in advanced technologies including 3D cell culture techniques.

Request Customization @ https://www.futuremarketinsights.com/customization-available/rep-gb-2843

Revenues in North America Continue to Surge

North America continued to spearhead the revenues of 3D cell culture market during the historical period and the status-quo is likely to continue in the future. In 2018, North America accounted for over two-fifths of the global 3D cell culture market revenues. The study estimates that regional revenues are expected to grow at 8% in 2019 over 2018.

Funding in research and development, especially in cancer research remains higher in the United States as compared to other developed countries. Europe also presents significant funding in R&D activities. The FMI study finds that over one-fourth of the 3D cell culture market revenues were accounted for the Europe region in 2018, of which a bulk of revenues come from Western European countries such as Germany, the UK, France, Italy and Spain.

Explore FMIs Extensive ongoing Coverage on Healthcare Domain

3D-Printed Prosthetic Implants Market - 3D-printed prosthetic implants are the identical body part which are designed using 3D printed technology. 3D scanning and printing is latest sci-fi technology which is used in various medical application such as designing the 3D-printed prosthetic implants.

3D Medical Implants Market - The three-dimensional (3D) medical implants are medical devices that offer aids in the medical reconstruction procedures of physical faults and abnormalities of the body. The 3D medical implants are commonly additively manufactured using 3D printers; which allow to develop complex geometry features and customized implants specific to the patients anatomy.

3D Surgical Microscope Systems Market - 3D surgical microscope system is an electrically or mechanically worked optical magnifying instrument intended for use in surgical settings for performing microsurgeries. The mix of focal points give stereoscopic vision, amplification going from 4x - 40x, and enlightened picture of the surgical zone.

About Future Market Insights (FMI)

Future Market Insights (FMI) is a leading provider of market intelligence and consulting services, serving clients in over 150 countries. FMI is headquartered in Dubai, and has delivery centers in the UK, U.S. and India. FMI's latest market research reports and industry analysis help businesses navigate challenges and make critical decisions with confidence and clarity amidst breakneck competition. Our customized and syndicated market research reports deliver actionable insights that drive sustainable growth. A team of expert-led analysts at FMI continuously tracks emerging trends and events in a broad range of industries to ensure that our clients prepare for the evolving needs of their consumers.

Contact:

Future Market Insights, 1602-6 Jumeirah Bay X2 Tower, Plot No: JLT-PH2-X2A, Jumeirah Lakes Towers, Dubai, United Arab Emirates For Sales Enquiries: sales@futuremarketinsights.com Website: https://www.futuremarketinsights.com/ Report: https://www.futuremarketinsights.com/reports/3d-cell-culture-market

Read more from the original source:
3D Cell Culture Market is Set to Experience a Significant Growth of 9.8% CAGR from 2022 To 2031 - Comprehensive Research Report By FMI - BioSpace

Lineage and Cancer Research UK Announce Completion of Patient Enrollment in Phase 1 Clinical Study of VAC2 for the Treatment of Non-small Cell Lung…

CARLSBAD, Calif.--(BUSINESS WIRE)-- Lineage Cell Therapeutics, Inc. (NYSE American and TASE: LCTX), a clinical-stage biotechnology company developing allogeneic cell therapies for unmet medical needs, announced today that Cancer Research UK recently completed patient enrollment in the ongoing Phase 1 clinical trial of VAC2, an allogeneic cancer vaccine product candidate, for the treatment of non-small cell lung cancer (NSCLC). Under the terms of an existing agreement, Cancer Research UK will complete the ongoing clinical trial and Lineage has now assumed responsibility for further clinical development of the VAC2 product candidate and any future development opportunities derived from the VAC platform.

We are pleased that Cancer Research UK has successfully completed patient enrollment in the VAC2 Phase 1 clinical study and overcame substantial challenges stemming from the COVID pandemic. We look forward to initial clinical results from this study being available later this year, stated Brian M. Culley, Lineage CEO. Clinical data previously collected by Cancer Research UK demonstrated peripheral immunogenicity in patients with NSCLC treated with VAC2, providing support to the underlying mechanism of using allogeneic dendritic cells to present tumor-associated antigens to the bodys immune system. Simultaneous with Cancer Research UK efforts to complete enrollment in the current study, the focus at Lineage has been on making improvements and modernizations to the VAC manufacturing process, an approach which we similarly employed in the development of OpRegen. We believe our focus on manufacturing will help prepare VAC2 for additional clinical trials and provide a competitive advantage for any future VAC programs which we advance, either alone or through alliances. With Cancer Research UK having completed enrollment of the current study, the team at Lineage also has begun work towards the submission of an Investigational New Drug Application for clinical testing of VAC2 in the U.S., which we anticipate submitting to the FDA later this year.

Dr. Nigel Blackburn, Director of Cancer Research UKs Centre for Drug Development, added: We are delighted to see that this innovative VAC2 program has reached such an important milestone in its development and are extremely proud to have played an important role in establishing its tolerability in lung cancer patients. We look forward to seeing Lineage advance VAC2 under their leadership in the future.

About VAC2

VAC2 is an allogeneic, or non-patient specific off-the-shelf, cancer vaccine product candidate designed to stimulate patient immune responses to an antigen commonly expressed in cancerous cells but not in normal adult cells. VAC2, which is produced from a pluripotent cell technology using a directed differentiation method, is comprised of a population of nonproliferating mature dendritic cells. As the most potent type of antigen presenting cell in the body, dendritic cells instruct the bodys immune system to attack and eliminate harmful pathogens and unwanted cells. Because the tumor antigen is loaded exogenously into the dendritic cells prior to administration, VAC2 is a platform technology that could be modified to carry selected antigens, including patient-specific tumor neo-antigens or viral antigens. VAC2 is currently being tested in a Phase 1 study in adult patients with NSCLC in the advanced and adjuvant settings (NCT03371485), conducted by Cancer Research UK.

About Cancer Research UKs Centre for Drug Development

Cancer Research UK has an impressive record of developing novel treatments for cancer. The Cancer Research UK Centre for Drug Development has been pioneering the development of new cancer treatments for 25 years, taking over 140 potential new anti-cancer agents into clinical trials in patients. It currently has a portfolio of 21 new anti-cancer agents in preclinical development, Phase I or early Phase II clinical trials. Six of these new agents have made it to market including temozolomide for brain cancer, abiraterone for prostate cancer and rucaparib for ovarian cancer. Two other drugs are in late development Phase III trials.

About Cancer Research UKs Commercial Partnerships Team

Cancer Research UK is the worlds leading cancer charity dedicated to saving lives through research. Cancer Research UKs specialist Commercial Partnerships Team works closely with leading international cancer scientists and their institutes to protect intellectual property arising from their research and to establish links with commercial partners. Cancer Research UKs commercial activity operates through Cancer Research Technology Ltd., a wholly owned subsidiary of Cancer Research UK. It is the legal entity which pursues drug discovery research in themed alliance partnerships and delivers varied commercial partnering arrangements.

About Cancer Research UK

For further information about Cancer Research UKs work or to find out how to support the charity, please call 0300 123 1022 or visit http://www.cancerresearchuk.org. Follow us on Twitter and Facebook.

About Lineage Cell Therapeutics, Inc.

Lineage Cell Therapeutics is a clinical-stage biotechnology company developing novel cell therapies for unmet medical needs. Lineages programs are based on its robust proprietary cell-based therapy platform and associated in-house development and manufacturing capabilities. With this platform Lineage develops and manufactures specialized, terminally differentiated human cells from its pluripotent and progenitor cell starting materials. These differentiated cells are developed to either replace or support cells that are dysfunctional or absent due to degenerative disease or traumatic injury or administered as a means of helping the body mount an effective immune response to cancer. Lineages clinical programs are in markets with billion dollar opportunities and include four allogeneic (off-the-shelf) product candidates: (i) OpRegen, a retinal pigment epithelium transplant therapy in Phase 1/2a development for the treatment of dry age-related macular degeneration, which is now being developed under a worldwide collaboration with Roche and Genentech, a member of the Roche Group; (ii) OPC1, an oligodendrocyte progenitor cell therapy in Phase 1/2a development for the treatment of acute spinal cord injuries; (iii) VAC2, a dendritic cell therapy produced from Lineages VAC technology platform for immuno-oncology and infectious disease, currently in Phase 1 clinical development for the treatment of non-small cell lung cancer and (iv) ANP1, an auditory neuronal progenitor cell therapy for the potential treatment of auditory neuropathy. For more information, please visit http://www.lineagecell.com or follow the Company on Twitter @LineageCell.

Forward-Looking Statements

Lineage cautions you that all statements, other than statements of historical facts, contained in this press release, are forward-looking statements. Forward-looking statements, in some cases, can be identified by terms such as believe, aim, may, will, estimate, continue, anticipate, design, intend, expect, could, can, plan, potential, predict, seek, should, would, contemplate, project, target, tend to, or the negative version of these words and similar expressions. Such statements include, but are not limited to, statements relating to the efficacy of using allogeneic dendritic cells to present tumor-associated antigens to the bodys immune system the collaboration and license agreement with Roche and Genentech and activities expected to occur thereunder, the broad potential for Lineages regenerative medicine platform as well as the VAC technology platform, and Lineages ability to expand the same; the projected timing of milestones of future studies, including their initiation and completion, projected manufacturing plans and improvements; the potential for Lineages investigational allogeneic cell therapies to generate clinical outcomes beyond the reach of traditional methods and provide safe and effective treatment for multiple, diverse serious or life threatening conditions. Forward-looking statements involve known and unknown risks, uncertainties and other factors that may cause Lineages actual results, performance or achievements to be materially different from future results, performance or achievements expressed or implied by the forward-looking statements in this press release, including, but not limited to, the risk that competing alternative therapies may adversely impact the commercial potential of OpRegen, which could materially adversely affect the milestone and royalty payments payable to Lineage under the collaboration and license agreement, the risk that Roche and Genentech may not be successful in completing further clinical trials for OpRegen and/or obtaining regulatory approval for OpRegen in any particular jurisdiction, the risk that Lineage might not succeed in developing products and technologies that are useful in medicine and demonstrate the requisite safety and efficacy to achieve regulatory approval in accordance with its projected timing, or at all; the risk that Lineage may not be able to manufacture sufficient clinical and, if approved, commercial quantities of its product candidates in accordance with current good manufacturing practice; the risks related to Lineages dependence on other third parties, and Lineages ability to establish and maintain its collaborations with these third parties; the risk that government-imposed bans or restrictions and religious, moral, and ethical concerns about the use of hES cells could prevent Lineage or its partners from developing and successfully marketing its stem cell product candidates; the risk that Lineages intellectual property may be insufficient to protect its products; the risk that the COVID-19 pandemic or geopolitical events may directly or indirectly cause significant delays in and substantially increase the cost of development of Lineages product candidates, as well as heighten other risks and uncertainties related to Lineages business and operations; risks and uncertainties inherent in Lineages business and other risks discussed in Lineages filings with the Securities and Exchange Commission (SEC). Lineages forward-looking statements are based upon its current expectations and involve assumptions that may never materialize or may prove to be incorrect. All forward-looking statements are expressly qualified in their entirety by these cautionary statements. Further information regarding these and other risks is included under the heading Risk Factors in Lineages periodic reports with the SEC, including Lineages most recent Annual Report on Form 10-K and Quarterly Report on Form 10-Q filed with the SEC and its other reports, which are available from the SECs website. You are cautioned not to place undue reliance on forward-looking statements, which speak only as of the date on which they were made. Lineage undertakes no obligation to update such statements to reflect events that occur or circumstances that exist after the date on which they were made, except as required by law.

View source version on businesswire.com: https://www.businesswire.com/news/home/20220413005356/en/

Read the original here:
Lineage and Cancer Research UK Announce Completion of Patient Enrollment in Phase 1 Clinical Study of VAC2 for the Treatment of Non-small Cell Lung...