Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.294
Filter
1.
Int J Pharm ; 659: 124292, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38823466

ABSTRACT

Camptothecin, a natural alkaloid, was first isolated from the bark and stem of the Camptotheca acuminate tree in China. It, along with its analogs, has demonstrated potent anti-cancer activity in preclinical studies, particularly against solid tumors such as lung, breast, ovarian, and colon cancer. Despite its promising anti-cancer activity, the application of camptothecin is limited due to its poor solubility, toxicity, and limited biodistribution. Nanotechnology-based drug delivery systems have been used to overcome limited bioavailability and ensure greater biodistribution after administration. Additionally, various drug delivery systems, particularly polymeric micelles, have been investigated to enhance the solubility, stability, and efficacy of camptothecin. Polymeric micelles offer a promising approach for the delivery of camptothecin. Polymeric micelles possess a core-shell structure, with a typical hydrophobic core, which exhibits a high capacity to incorporate hydrophobic drugs. The structure of polymeric micelles can be engineered to have a high drug loading capacity, thereby enabling them to carry a large amount of hydrophobic drug within their core. The shell portion of polymeric micelles is composed of hydrophilic polymers Furthermore, the hydrophilic segment of polymeric micelles plays an important role in protecting against the reticuloendothelial system (RES). This review provides a discussion on recent research and developments in the delivery of camptothecin using polymeric micelles for the treatment of cancers.


Subject(s)
Antineoplastic Agents, Phytogenic , Camptothecin , Drug Delivery Systems , Micelles , Polymers , Camptothecin/administration & dosage , Camptothecin/chemistry , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Humans , Polymers/chemistry , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Drug Delivery Systems/methods , Neoplasms/drug therapy , Drug Carriers/chemistry , Solubility , Tissue Distribution , Hydrophobic and Hydrophilic Interactions
2.
Carbohydr Polym ; 339: 122257, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823923

ABSTRACT

Traditional solid phase extraction (SPE) suffers from a lack of specific adsorption. To overcome this problem, a combination of adsorption method and molecular imprinting technology by polydopamine modification was proposed to realize specific recognition of target compounds in SPE, which is of great significance to improve the separation efficiency of SPE. Cellulose hydrogel beads were prepared by dual cross-linking curing method and modified with polydopamine to make them hydrophilic and biocompatible. Subsequently, cellulose hydrogel-based molecularly imprinted beads (MIBs) were synthesized by surface molecular imprinting technology and used as novel column fillers in SPE to achieve efficient adsorption (34.16 mg·g-1) with specific selectivity towards camptothecin (CPT) in 120 min. The simulation and NMR analysis revealed that recognition mechanism of MIBs involved hydrogen bond interactions and Van der Waals effect. The MIBs were successful used in separating CPT from Camptotheca acuminata fruits, exhibiting impressive adsorption capacity (1.19 mg·g-1) and efficient recovery of CPT (81.54 %). Thus, an environmentally friendly column filler for SPE was developed, offering a promising avenue for utilizing cellulose-based materials in the selective separation of natural products.


Subject(s)
Camptothecin , Cellulose , Hydrogels , Molecular Imprinting , Solid Phase Extraction , Camptothecin/chemistry , Camptothecin/isolation & purification , Cellulose/chemistry , Adsorption , Molecular Imprinting/methods , Hydrogels/chemistry , Solid Phase Extraction/methods , Camptotheca/chemistry , Polymers/chemistry , Hydrophobic and Hydrophilic Interactions , Indoles/chemistry , Fruit/chemistry
3.
Bioorg Chem ; 148: 107436, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735265

ABSTRACT

BACKGROUND: Camptothecin (CPT), a pentacyclic alkaloid with antitumor properties, is derived from the Camptotheca acuminata. Topotecan and irinotecan (CPT derivatives) were first approved by the Food and Drug Administration for cancer treatment over 25 years ago and remain key anticancer drugs today. However, their use is often limited by clinical toxicity. Despite extensive development efforts, many of these derivatives have not succeeded clinically, particularly in their effectiveness against pancreatic cancer which remains modest. AIM OF THE STUDY: This study aimed to evaluate the therapeutic activity of FLQY2, a CPT derivative synthesized in our laboratory, against pancreatic cancer, comparing its efficacy and mechanism of action with those of established clinical drugs. METHODS: The cytotoxic effects of FLQY2 on cancer cells were assessed using an MTT assay. Patient-derived organoid (PDO) models were employed to compare the sensitivity of FLQY2 to existing clinical drugs across various cancers. The impact of FLQY2 on apoptosis and cell cycle arrest in Mia Paca-2 pancreatic cancer cells was examined through flow cytometry. Transcriptomic and proteomic analyses were conducted to explore the underlying mechanisms of FLQY2's antitumor activity. Western blotting was used to determine the levels of proteins regulated by FLQY2. Additionally, the antitumor efficacy of FLQY2 in vivo was evaluated in a pancreatic cancer xenograft model. RESULTS: FLQY2 demonstrated (1) potent cytotoxicity; (2) superior tumor-suppressive activity in PDO models compared to current clinical drugs such as gemcitabine, 5-fluorouracil, cisplatin, paclitaxel, ivosidenib, infinitinib, and lenvatinib; (3) significantly greater tumor inhibition than paclitaxel liposomes in a pancreatic cancer xenograft model; (4) robust antitumor effects, closely associated with the inhibition of the TOP I and PDK1/AKT/mTOR signaling pathways. In vitro studies revealed that FLQY2 inhibited cell proliferation, colony formation, induced apoptosis, and caused cell cycle arrest at nanomolar concentrations. Furthermore, the combination of FLQY2 and gemcitabine exhibited significant inhibitory and synergistic effects. CONCLUSION: The study confirmed the involvement of topoisomerase I and the PDK1/AKT/mTOR pathways in mediating the antitumor activity of FLQY2 in treating Mia Paca-2 pancreatic cancer. Therefore, FLQY2 has potential as a novel therapeutic option for patients with pancreatic cancer.


Subject(s)
Antineoplastic Agents , Apoptosis , Camptothecin , Cell Proliferation , Drug Screening Assays, Antitumor , Pancreatic Neoplasms , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Camptothecin/pharmacology , Camptothecin/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Animals , Mice , Apoptosis/drug effects , Structure-Activity Relationship , Molecular Structure , Dose-Response Relationship, Drug , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Mice, Nude , Tumor Cells, Cultured , Cell Line, Tumor
4.
ACS Nano ; 18(20): 12933-12944, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38712906

ABSTRACT

Efficient tumor-targeted drug delivery is still a challenging and currently unbreakable bottleneck in chemotherapy for tumors. Nanomedicines based on passive or active targeting strategy have not yet achieved convincing chemotherapeutic benefits in the clinic due to the tumor heterogeneity. Inspired by the efficient inflammatory-cell recruitment to acute clots, we constructed a two-component nanosystem, which is composed of an RGD-modified pyropheophorbide-a (Ppa) micelle (PPRM) that mediates the tumor vascular-targeted photodynamic reaction to activate local coagulation and subsequently transmits the coagulation signals to the circulating clot-targeted CREKA peptide-modified camptothecin (CPT)-loaded nanodiscs (CCNDs) for amplifying tumor targeting. PPRM could effectively bind with the tumor vasculature and induce sufficient local thrombus by a photodynamic reaction. Local photodynamic reaction-induced tumor target amplification greatly increased the tumor accumulation of CCND by 4.2 times, thus significantly enhancing the chemotherapeutic efficacy in the 4T1 breast tumor model. In other words, this study provides a powerful platform to amplify tumor-specific drug delivery by taking advantage of the efficient crosstalk between the PPRM-activated coagulation cascade and clot-targeted CCND.


Subject(s)
Chlorophyll , Nanoparticles , Photochemotherapy , Animals , Nanoparticles/chemistry , Mice , Chlorophyll/analogs & derivatives , Chlorophyll/chemistry , Chlorophyll/pharmacology , Drug Delivery Systems , Female , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Camptothecin/chemistry , Camptothecin/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/administration & dosage , Micelles , Mice, Inbred BALB C , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Oligopeptides/chemistry , Oligopeptides/pharmacology
5.
Biomaterials ; 309: 122606, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38776593

ABSTRACT

Carbon monoxide (CO) has emerged as a potential antitumor agent by inducing the dysfunction of mitochondria and the apoptosis of cancer cells. However, it remains challenging to deliver appropriate amount of CO into tumor to ensure efficient tumor growth suppression with minimum side effects. Herein we developed a CO prodrug-loaded nanomedicine based on the self-assembly of camptothecin (CPT) polyprodrug amphiphiles. The polyprodrug nanoparticles readily dissociate upon exposure to endogenous H2O2 in the tumor, resulting in rapid release of CPT and generation of high-energy intermediate dioxetanedione. The latter can transfer the energy to neighboring CO prodrugs to activate CO production by chemiexcitation, while CPT promotes the generation of H2O2 in tumors, which in turn facilitates cascade CPT and CO release. As a result, the polyprodrug nanoparticles display remarkable tumor suppression in both subcutaneous and orthotopic breast tumor-bearing mice owing to the self-augmented CPT release and CO generation. In addition, no obvious systemic toxicity was observed in mice treated with the metal-free CO prodrug-loaded nanomedicine, suggesting the good biocompatibility of the polyprodrug nanoparticles. Our work provides new insights into the design and construction of polyprodrug nanomedicines for synergistic chemo/gas therapy.


Subject(s)
Camptothecin , Carbon Monoxide , Nanomedicine , Nanoparticles , Prodrugs , Animals , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/therapeutic use , Nanomedicine/methods , Camptothecin/pharmacology , Camptothecin/therapeutic use , Camptothecin/administration & dosage , Camptothecin/chemistry , Female , Humans , Carbon Monoxide/chemistry , Nanoparticles/chemistry , Cell Line, Tumor , Mice, Inbred BALB C , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Hydrogen Peroxide/chemistry , Mice, Nude
6.
Sci Prog ; 107(2): 368504241253675, 2024.
Article in English | MEDLINE | ID: mdl-38807531

ABSTRACT

Camptothecin (CPT) is an important alkaloid used for anticancer treatment. It is mainly produced by two endangered and overharvested Camptotheca acuminata and Nothapodytes nimmoniana plants. Endophytic fungi are promising alternative sources for CPT production. In the present study, fungi residing within explants of Ixora chinensis were isolated and their CPT-producing capability of their endophytes was verified via thin-layer chromatography, high-performance liquid chromatography, liquid chromatography/high resolution mass spectrometry, and nuclear magnetic resonance analyses and compared with standards. In addition, MTT and sulforhodamine B assays were selected to test the anticancer effect. The endophytic fungi collection of 62 isolates were assigned to 11 genera, with four common genera (Diaporthe, Phyllosticta, Colletotrichum, and Phomopsis) and seven less common genera (Penicillium, Botryosphaeria, Fusarium, Pestalotiopsis, Aspergillus, and Didymella). Moreover, the anticancer activity of extracts was assessed against human lung carcinoma (A549). Among eight potential extracts, only Penicillium sp. I3R2 was found to be a source of CPT, while the remaining seven extracts have not been discovered potential secondary compounds. Thus, other prominent endophytic fungi might be potential candidates of phytochemicals with anticancer properties.


Subject(s)
Antineoplastic Agents , Camptothecin , Endophytes , Fungi , Humans , Camptothecin/pharmacology , Camptothecin/chemistry , Camptothecin/biosynthesis , Endophytes/metabolism , Endophytes/isolation & purification , Endophytes/chemistry , Fungi/isolation & purification , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , A549 Cells , Cell Line, Tumor
7.
Int J Pharm ; 659: 124283, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38810933

ABSTRACT

The clinical application of 7-ethyl hydroxy-camptothecin (SN-38) maintains challenges not only due to its poor solubility and stability but also the lack of effective carriers to actively deliver SN-38 to deep tumor sites. Although SN-38-based nanomedicines could improve the solubility and stability from different aspects, the tumor targeting efficiency remains very low. Leveraging the hypoxic taxis of bifidobacteria bifidum (B. bifi) to the deep tumor area, we report SN-38-based nanomedicines-engineered bifidobacterial complexes for effective tumor-targeted delivery. Firstly, SN-38 was covalently coupled with poly-L-glutamic acid (L-PGA) and obtained soluble polymeric prodrug L-PGA-SN38 to improve its solubility and stability. To prolong the drug release, L-PGA-SN38 was mildly complexed with chitosan to form nanomedicines, and nanomedicines engineered B. bifi were further elaborated via electrostatic interaction of the excess of cationic chitosan shell from nanomedicines and anionic teichoic acid from B. bifi. The engineered B. bifi complexes inherited the bioactivity of native B. bifi and exhibited distinctly enhanced accumulation at the tumor site. More importantly, significantly elevated anti-tumor efficacy was achieved after the treatment of CS-L-PGA-SN38 NPs/B. bifi complexes, with favorable tumor suppression up to 80%. Such a B. bifi-mediated delivery system offers a promising platform for effective drug delivery and enhanced drug accumulation in the hypoxia deep tumor with superior anti-tumor efficacy.


Subject(s)
Chitosan , Colorectal Neoplasms , Irinotecan , Nanomedicine , Polyglutamic Acid , Irinotecan/administration & dosage , Irinotecan/pharmacology , Chitosan/chemistry , Colorectal Neoplasms/drug therapy , Animals , Polyglutamic Acid/chemistry , Polyglutamic Acid/analogs & derivatives , Humans , Nanomedicine/methods , Drug Liberation , Drug Carriers/chemistry , Drug Delivery Systems , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology , Mice , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/chemistry , Camptothecin/pharmacology , Mice, Inbred BALB C , Cell Line, Tumor , Bifidobacterium bifidum , Mice, Nude , Female
8.
ACS Appl Bio Mater ; 7(5): 3202-3214, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38651918

ABSTRACT

The combination of small-interfering RNA (siRNA)-mediated gene silencing and chemotherapeutic agents for lung cancer treatment has attracted widespread attention in terms of a greater therapeutic effect, minimization of systemic toxicity, and inhibition of multiple drug resistance (MDR). In this work, three amphiphiles, CBN1-CBN3, were first designed and synthesized as a camptothecin (CPT) conjugate and gene condensation agents by the combination of CPT prodrugs and di(triazole-[12]aneN3) through the ROS-responsive phenylborate ester and different lengths of alkyl chains (with 6, 9, 12 carbon chains for CBN1-CBN3, respectively). CBN1-CBN3 were able to be self-assembled into liposomes with an average diameter in the range of 320-240 nm, showing the ability to effectively condense siRNA. Among them, CBN2, with a nine-carbon alkyl chain, displayed the best anticancer efficiency in A549 cells. In order to give nanomedicines a stealth property and PEGylation/dePEGylation transition, a GSH-responsive PEGylated TPE derivative containing a disulfide linkage (TSP) was further designed and prepared. A combination of CBN2/siRNA complexes and DOPE with TSP resulted in GSH/ROS dual-responsive lipid-polymer hybrid nanoparticles (CBN2-DP/siRNA NPs). In present GSH and H2O2, CBN2-DP/siRNA NPs were decomposed, resulting in the controlled release of CPT drug and siRNA. In vitro, CBN2-DP/siPHB1 NPs showed the best anticancer activity for suppression of about 75% of A549 cell proliferation in a serum medium. The stability of CBN2-DP/siRNA NPs was significantly prolonged in blood circulation, and they showed effective accumulation in the A549 tumor site through an enhanced permeability and retention (EPR) effect. In vivo, CBN2-DP/siPHB1 NPs demonstrated enhanced synergistic cancer therapy efficacy and tumor inhibition as high as 71.2%. This work provided a strategy for preparing lipid-polymer hybrid NPs with GSH/ROS dual-responsive properties and an intriguing method for lung cancer therapy.


Subject(s)
Biocompatible Materials , Camptothecin , Cell Proliferation , Drug Screening Assays, Antitumor , Lung Neoplasms , Nanoparticles , RNA, Small Interfering , Reactive Oxygen Species , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , RNA, Small Interfering/chemistry , Camptothecin/chemistry , Camptothecin/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemical synthesis , Cell Proliferation/drug effects , Materials Testing , Glutathione/chemistry , Glutathione/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , A549 Cells , Particle Size , Lipids/chemistry , Molecular Structure , Animals , Cell Survival/drug effects , Mice , Prohibitins
9.
J Control Release ; 369: 622-629, 2024 May.
Article in English | MEDLINE | ID: mdl-38604383

ABSTRACT

Enhancing the delivery and release efficiency of hydroxyl agents, constrained by high pKa values and issues of release rate or unstable linkage, is a critical challenge. To address this, a self-immolative linker, composed of a modifiable p-hydroxybenzyl ether and a fast cyclization adapter (N-(ortho-hydroxyphenyl)-N-methylcarbamate) was strategically designed, for the synthesis of prodrugs. The innovative linker not only provides a side chain modification but also facilitates the rapid release of the active payloads, thereby enabling precise drug delivery. Particularly, five prodrug model compounds (J1, J2, J3, J5 and J6) were synthesized to evaluate the release rates by using ß-glucuronic acid as trigger and five hydroxyl compounds as model payloads. Significantly, all prodrug model compounds could efficiently release the hydroxyl payloads under the action of ß-glucuronidase, validating the robustness of the linker. And then, to assess the drug delivery and release efficiency using endogenous albumin as a transport vehicle, J1148, a SN38 prodrug modified with maleimide side chain was synthesized. Results demonstrated that J1148 covalently bound to plasma albumin through in situ Michael addition, effectively targeting the tumor microenvironment. Activated by ß-glucuronidase, J1148 underwent a classical 1, 6-elimination, followed by rapid cyclization of the adapter, thereby releasing SN38. Impressively, J1148 showed excellent therapeutic efficacy against human colonic cancer xenograft model, leading to a significant reduction or even disappearance of tumors (3/6 of mice cured). These findings underscore the potential of the designed linker in the delivery system of hydroxyl agents, positioning it at the forefront of advancements in drug delivery technology.


Subject(s)
Drug Delivery Systems , Irinotecan , Prodrugs , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Animals , Humans , Irinotecan/administration & dosage , Irinotecan/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Camptothecin/chemistry , Drug Liberation , Mice, Nude , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Female , Mice , Albumins/administration & dosage , Albumins/chemistry , Glucuronidase/metabolism , Mice, Inbred BALB C
10.
Bioorg Chem ; 147: 107371, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643564

ABSTRACT

Due to the strong selectivity and permeability of tumor tissue, anti-cancer peptide-drug conjugates (PDCs) can accumulate high concentration of toxic payloads at the target, effectively killing tumor cells. This approach holds great promise for tumor-targeted treatment. In our previous study, we identified the optimal peptide P1 (NPNWGRSWYNQRFK) targeting HER2 from pertuzumab, a monoclonal antibody that blocks the HER2 signaling pathway. Here, a series of PDCs were constructed through connecting P1 and CPT with different linkers. Among these, Z8 emerged as the optimal compound, demonstrating good antitumor activity and targeting ability in biological activity tests. Z8 exhibited IC50 values of 1.04 ± 0.24 µM and 1.91 ± 0.71 µM against HER2-positive SK-BR-3 and NCI-N87 cells, respectively. Moreover, superior antitumor activity and higher biosafety of Z8 were observed compared to the positive control CPT in vivo, suggesting a novel idea for the construction of PDCs.


Subject(s)
Antineoplastic Agents , Camptothecin , Cell Proliferation , Drug Screening Assays, Antitumor , Peptides , Receptor, ErbB-2 , Humans , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Camptothecin/pharmacology , Camptothecin/chemistry , Structure-Activity Relationship , Animals , Cell Proliferation/drug effects , Peptides/chemistry , Peptides/pharmacology , Peptides/chemical synthesis , Molecular Structure , Dose-Response Relationship, Drug , Mice , Drug Discovery , Cell Line, Tumor , Female , Mice, Inbred BALB C , Mice, Nude
11.
Colloids Surf B Biointerfaces ; 238: 113906, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615388

ABSTRACT

Combination chemotherapy has been recognized as a more powerful strategy for tumor treatment rather than the single chemotherapy. However, the interactive mechanism of the two hydrophobic chemotherapeutic drugs has not been explored by now. Aiming for a better synergistic effect, such interactive mechanism was investigated in the present work, by designing CPT@DOX-DPUTEA-PEG nanomedicine with encapsulated camptothecin (CPT) and conjugated doxorubicin (DOX). The synergistic controlled drug release effect was found for the two drugs loaded on the different sites of the dendritic polyurethane core. Synergism was achieved on the HepG2 cells with a combination index (CI) of 0.58 in the in vitro cellular experiments. The results demonstrated the promising application of the unimolecular micelles-based nanomedicine with independently loading of two hydrophobic chemotherapeutic drugs.


Subject(s)
Camptothecin , Doxorubicin , Drug Liberation , Micelles , Prodrugs , Doxorubicin/pharmacology , Doxorubicin/chemistry , Camptothecin/pharmacology , Camptothecin/chemistry , Humans , Hydrogen-Ion Concentration , Hep G2 Cells , Prodrugs/chemistry , Prodrugs/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Polymers/chemistry , Cell Survival/drug effects , Dendrimers/chemistry , Drug Delivery Systems , Drug Synergism , Polyethylene Glycols/chemistry
12.
J Colloid Interface Sci ; 667: 119-127, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38631250

ABSTRACT

The current state of drug delivery systems allows for the resolution of specific issues like inadequate solubility, limited targeting capabilities, and complex preparation processes, requiring tailored designs for different drugs. Yet, the major challenge in clinical application lies in surmounting these obstacles with a universal carrier that is effective for a variety of anticancer drugs. Herein, with the help of computer simulation, we rationally design ultrashort peptides GY and CCYRGD, which can co-assemble with hydrophobic anticancer drugs into nanoparticles with enhanced solubility, targeting ability and anticancer efficacy. Taking 7-ethyl-10-hydroxy camptothecin (SN38) as a model anticancer drug, the co-assembled SN38-GY-CCYRGD nanoparticles significantly enhance the water solubility of SN38 by more than three orders of magnitude. The as-prepared nanoparticles can effectively kill cancer cells, e.g., human small cell lung cancer (A549) cells with a notable cell mortality rate of 71%. Mice experimental results demonstrate the nanoparticles' efficient targeting capability, marked reducing the toxicity to normal tissues while improving antitumor efficacy. This work presents a novel drug delivery method, integrating effective, targeted, and safe strategies into a comprehensive carrier system, designed for the administration of hydrophobic anticancer drugs.


Subject(s)
Antineoplastic Agents , Hydrophobic and Hydrophilic Interactions , Nanoparticles , Peptides , Solubility , Humans , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Mice , Nanoparticles/chemistry , Peptides/chemistry , Peptides/pharmacology , Irinotecan/pharmacology , Irinotecan/chemistry , A549 Cells , Drug Carriers/chemistry , Cell Survival/drug effects , Particle Size , Drug Delivery Systems , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects , Mice, Inbred BALB C , Surface Properties , Camptothecin/chemistry , Camptothecin/pharmacology , Camptothecin/administration & dosage
13.
Mol Pharm ; 21(5): 2327-2339, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38576375

ABSTRACT

In the present study, we investigated the role of lipid composition of camptothecin (CPT)-loaded liposomes (CPT-Lips) to adjust their residence time, drug distribution, and therefore the toxicities and antitumor activity. The CPT was loaded into liposomes using a click drug loading method, which utilized liposomes preloaded with GSH and then exposed to CPT-maleimide. The method produced CPT-Lips with a high encapsulation efficiency (>95%) and sustained drug release. It is shown that the residence times of CPT-Lips in the body were highly dependent on lipid compositions with an order of non-PEGylated liposomes of unsaturated lipids < non-PEGylated liposomes of saturated lipids < PEGylated liposomes of saturated lipids. Interestingly, the fast clearance of CPT-Lips resulted in significantly decreased toxicities but did not cause a significant decrease in their in vivo antitumor activity. These results suggested that the lipid composition could effectively adjust the residence time of CPT-Lips in the body and further optimize their therapeutic index, which would guide the development of a liposomal formulation of CPT.


Subject(s)
Camptothecin , Lipids , Liposomes , Camptothecin/chemistry , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Liposomes/chemistry , Animals , Mice , Lipids/chemistry , Humans , Drug Liberation , Drug Delivery Systems/methods , Polyethylene Glycols/chemistry , Cell Line, Tumor , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Female , Click Chemistry/methods , Mice, Inbred BALB C
14.
Adv Mater ; 36(23): e2312153, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38444205

ABSTRACT

The presence of bacteria in tumor results in chemotherapeutic drug resistance and weakens the immune response in colorectal cancer. To overcome bacterium-induced chemotherapeutic drug resistance and potentiate antitumor immunity, herein a novel molecule Biotin-Lys(SA-Cip-OH)-Lys(SA-CPT)-Phe-Phe-Nap (Biotin-Cip-CPT-Nap) is rationally designed containing four functional motifs (i.e., a biotin motif for targeting, Phe-Phe(-Nap) motif for self-assembly, ciprofloxacin derivative (Cip-OH) motif for antibacterial effect, and camptothecin (CPT) motif for chemotherapy). Using the designed molecule, a novel strategy of intracellular enzymatic nanofiber formation and synergistic antibacterium-enhanced chemotherapy and immunotherapy is achieved. Under endocytosis mediated by highly expressed biotin receptor in colorectal cancer cell membrane and the catalysis of highly expressed carboxylesterase in the cytoplasm, this novel molecule can be transformed into Biotin-Nap, which self-assembled into nanofibers. Meanwhile, antibiotic Cip-OH and chemotherapeutic drug CPT are released, overcoming bacterium-induced drug resistance and enhancing the therapeutic efficacy of immunotherapy towards colorectal cancer. This work offers a feasible strategy for the design of novel multifunctional prodrugs to improve the efficiency of colorectal cancer treatment.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Animals , Immunotherapy , Peptides/chemistry , Peptides/pharmacology , Camptothecin/pharmacology , Camptothecin/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mice , Nanofibers/chemistry , Ciprofloxacin/pharmacology , Ciprofloxacin/chemistry , Drug Liberation , Biotin/chemistry
15.
Mol Cancer Ther ; 23(5): 606-618, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38354417

ABSTRACT

In recent years, the field of antibody drug conjugates (ADC) has seen a resurgence, largely driven by the clinical benefit observed in patients treated with ADCs incorporating camptothecin-based topoisomerase I inhibitor payloads. Herein, we present the development of a novel camptothecin ZD06519 (FD1), which has been specifically designed for its application as an ADC payload. A panel of camptothecin analogs with different substituents at the C-7 and C-10 positions of the camptothecin core was prepared and tested in vitro. Selected compounds spanning a range of potency and hydrophilicity were elaborated into drug-linkers, conjugated to trastuzumab, and evaluated in vitro and in vivo. ZD06519 was selected on the basis of its favorable properties as a free molecule and as an antibody conjugate, which include moderate free payload potency (∼1 nmol/L), low hydrophobicity, strong bystander activity, robust plasma stability, and high-monomeric ADC content. When conjugated to different antibodies using a clinically validated MC-GGFG-based linker, ZD06519 demonstrated impressive efficacy in multiple cell line-derived xenograft models and noteworthy tolerability in healthy mice, rats, and non-human primates.


Subject(s)
Camptothecin , Immunoconjugates , Xenograft Model Antitumor Assays , Camptothecin/pharmacology , Camptothecin/chemistry , Immunoconjugates/pharmacology , Immunoconjugates/chemistry , Animals , Humans , Mice , Cell Line, Tumor , Drug Design , Female , Rats
16.
Adv Healthc Mater ; 13(14): e2303626, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38387885

ABSTRACT

Immunotherapy has emerged as an innovative strategy with the potential to improve outcomes in cancer patients. Recent evidence indicates that radiation-induced DNA damage can activate the cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway to enhance the antitumor immune response. Even so, only a small fraction of patients currently benefits from radioimmunotherapy due to the radioresistance and the inadequate activation of the cGAS-STING pathway. Herein, this work integrates hafnium oxide (HfO2) nanoparticles (radiosensitizer) and 7-Ethyl-10-hydroxycamptothecin (SN38, chemotherapy drug, STING agonist) into a polydopamine (PDA)-coated core-shell nanoplatform (HfO2@PDA/Fe/SN38) to achieve synergistic chemoradiotherapy and immunotherapy. The co-delivery of HfO2/SN38 greatly enhances radiotherapy efficacy by effectively activating the cGAS-STING pathway, which then triggers dendritic cells maturation and CD8+ T cells recruitment. Consequently, the growth of both primary and abscopal tumors in tumor-bearing mice is efficiently inhibited. Moreover, the HfO2@PDA/Fe/SN38 complexes exhibit favorable magnetic resonance imaging (MRI)/photoacoustic (PA) bimodal molecular imaging properties. In summary, these developed multifunctional complexes have the potential to intensify immune activation to realize simultaneous cancer Radio/Chemo/Immunotherapy for clinical translation.


Subject(s)
Immunotherapy , Membrane Proteins , Nanoparticles , Nucleotidyltransferases , Animals , Nucleotidyltransferases/metabolism , Membrane Proteins/metabolism , Mice , Immunotherapy/methods , Nanoparticles/chemistry , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , Humans , Camptothecin/pharmacology , Camptothecin/chemistry , Camptothecin/analogs & derivatives , Molecular Imaging/methods , Polymers/chemistry , Neoplasms/therapy , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Signal Transduction/drug effects , Indoles/chemistry , Indoles/pharmacology , Female
17.
Phytochem Anal ; 35(4): 825-839, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38351290

ABSTRACT

INTRODUCTION: Efficient extraction of camptothecin (CPT), an anticancer agent from the commercial source Nothapodytes nimmoniana (J. Graham) Mabb in India, is of paramount importance. CPT is present in the highest concentration in the stem portion, and the stem can be readily harvested without uprooting the plant. The fluorescence microscopy mapping of the bark matrix for CPT revealed its presence in a free form within both the outer (epidermal and cortical tissues) and inner (xylem and phloem tissues) sections. The bark matrix primarily consists of cellulose, hemicellulose, and lignin, rendering it woody, rigid, and resistant to efficient solvent penetration for CPT extraction. We proposed a hypothesis that subjecting it to disruption through treatment with hydrolytic enzymes like cellulase and xylanase could enhance solvent diffusion, thereby enabling a swift and effective extraction of CPT. OBJECTIVE: The present study was aimed at enzyme-assisted extraction, using cellulase and xylanase for hydrolytic disruption of the cells to readily access CPT from the stem of the plant N. nimmoniana (J. Graham) Mabb. METHODOLOGY: The hydrolytic cell disruption of ground powder from the stem bark was studied using cellulase and xylanase enzymes. The enzymatically pretreated stem bark powder was subsequently recovered by filtration, dried, and subjected to extraction with methanol to isolate CPT. This process was optimised through a Box-Behnken design, employing a one-factor-at-a-time approach to assess parameters such as enzyme concentration (2-10% w/w), pH (3-7), incubation time (6-24 h), and solid-to-solvent ratio (1:30-1:70 g/mL). CPT was characterised using proton nuclear magnetic resonance (1H-NMR) and Fourier transform infrared (FTIR) spectra, and a high-performance liquid chromatography (HPLC) method was developed for quantification. RESULTS: The cellulase and xylanase treatment resulted in the highest yields of 0.285% w/w and 0.343% w/w, with efficiencies of 67% and 81%, respectively, achieved in a significantly shorter time compared to the untreated material, which yielded 0.18% with an efficiency of only 42%. Extraction by utilising the predicted optimised process parameters, a nearly two-fold increase in the yield, was observed for xylanase, with incubation and solvent extraction times set at 16 and 2 h, respectively. Scanning electron microscopy (SEM) images of the spent material indicated perforations attributed to enzymatic action, suggesting that this could be a primary factor contributing to the enhanced extraction. CONCLUSION: Enzyme-mediated hydrolytic cell disruption could be a potential approach for efficient and rapid isolation of CPT from the bark of N. nimmoniana.


Subject(s)
Camptothecin , Camptothecin/chemistry , Cellulase/chemistry , Cellulase/metabolism , Endo-1,4-beta Xylanases/metabolism , Endo-1,4-beta Xylanases/chemistry , Plant Bark/chemistry , Chromatography, High Pressure Liquid/methods
18.
Biomolecules ; 14(2)2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38397391

ABSTRACT

Pro-drugs, which ideally release their active compound only at the site of action, i.e., in a cancer cell, are a promising approach towards an increased specificity and hence reduced side effects in chemotherapy. A popular form of pro-drugs is esters, which are activated upon their hydrolysis. Since carboxylesterases that catalyse such a hydrolysis reaction are also abundant in normal tissue, it is of great interest whether a putative pro-drug is a probable substrate of such an enzyme and hence bears the danger of being activated not just in the target environment, i.e., in cancer cells. In this work, we study the binding mode of carboxylesters of the drug molecule camptothecin, which is an inhibitor of topoisomerase I, of varying size to human carboxylesterase 2 (HCE2) by molecular docking and molecular dynamics simulations. A comparison to irinotecan, known to be a substrate of HCE2, shows that all three pro-drugs analysed in this work can bind to the HCE2 protein, but not in a pose that is well suited for subsequent hydrolysis. Our data suggest, moreover, that for the irinotecan substrate, a reactant-competent pose is stabilised once the initial proton transfer from the putative nucleophile Ser202 to the His431 of the catalytic triad has already occurred. Our simulation work also shows that it is important to go beyond the static models obtained from molecular docking and include the flexibility of enzyme-ligand complexes in solvents and at a finite temperature. Under such conditions, the pro-drugs studied in this work are unlikely to be hydrolysed by the HCE2 enzyme, indicating a low risk of undesired drug release in normal tissue.


Subject(s)
Camptothecin , Carboxylesterase , Irinotecan , Prodrugs , Humans , Camptothecin/chemistry , Carboxylesterase/chemistry , Irinotecan/chemistry , Molecular Docking Simulation , Prodrugs/chemistry , Protein Binding
19.
J AOAC Int ; 107(3): 387-395, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38407421

ABSTRACT

BACKGROUND: Camptothecin is a potent anticancer drug used for the treatment of various cancers. OBJECTIVE: The goal of this research investigation was to develop and validate a new stability-indicating HPLC technique for the quantitative assessment of camptothecin in in-house developed mesoporous silica nanoparticles, a novel nanoformulation matrix for the treatment of cancer. METHOD: The Waters Inertsil® HPLC column (C18) was used for the chromatographic separation, with a flow rate of 1 mL/min, a column oven temperature of 40°C, an injection volume of 10 µL, a detection wavelength of 216 nm, and a 10 min runtime overall. An isocratic blend of phosphate buffer (10 mM, pH7.0) and acetonitrile (60:40, v/v) served as the mobile phase. Various stress conditions including acid, alkali, oxidative, photolytic, thermal, and humidity environments were tested for the quantitative estimation of the camptothecin through the proposed method. RESULTS: The results demonstrated that the proposed method is specific (peak purity ≥0.999), accurate (99.69-100.64% w/w), precise (RSD, % <2.0), and sensitive (LOD-0.17 µg and LOQ-0.56 µg) in accordance with ICH guideline Q2 (R1). Any unidentified degradation products did not interfere with the drug's estimation. Furthermore, the current method of analysis has eliminated any excipient interference from the matrix effect caused by the numerous excipients of the formulation matrix. CONCLUSIONS: To quantify camptothecin for routine assay purposes, this research work offers a novel and straightforward HPLC methodology with optimized chromatographic parameters, contributing to the research and development community while ensuring an appropriate and efficient use of the drug through a variety of nanoformulation for cancer treatment. HIGHLIGHTS: The stability-indicating HPLC method was found to be specific and suitable for routine analysis of camptothecin. The absence of any interference from excipients was confirmed by forced degradation studies.


Subject(s)
Camptothecin , Nanoparticles , Silicon Dioxide , Chromatography, High Pressure Liquid/methods , Camptothecin/analysis , Camptothecin/chemistry , Silicon Dioxide/chemistry , Nanoparticles/chemistry , Porosity , Drug Stability
20.
Angew Chem Int Ed Engl ; 63(18): e202314143, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38179812

ABSTRACT

Drug safety and efficacy due to premature release into the bloodstream and poor biodistribution remains a problem despite seminal advances in this area. To circumvent these limitations, we report drug cyclization based on dynamic covalent linkages to devise a dual lock for the small-molecule anticancer drug, camptothecin (CPT). Drug activity is "locked" within the cyclic structure by the redox responsive disulfide and pH-responsive boronic acid-salicylhydroxamate and turns on only in the presence of acidic pH, reactive oxygen species and glutathione through traceless release. Notably, the dual-responsive CPT is more active (100-fold) than the non-cleavable (permanently closed) analogue. We further include a bioorthogonal handle in the backbone for functionalization to generate cyclic-locked, cell-targeting peptide- and protein-CPTs, for targeted delivery of the drug and traceless release in triple negative metastatic breast cancer cells to inhibit cell growth at low nanomolar concentrations.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Camptothecin/chemistry , Tissue Distribution , Antineoplastic Agents/chemistry , Micelles , Proteins , Drug Delivery Systems , Nanoparticles/chemistry , Drug Liberation , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL
...