1
|
Martin JD, Miyazaki T, Cabral H. Remodeling tumor microenvironment with nanomedicines. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1730. [PMID: 34124849 DOI: 10.1002/wnan.1730] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022]
Abstract
The tumor microenvironment (TME) has been recognized as a major contributor to cancer malignancy and therapeutic resistance. Thus, strategies directed to re-engineer the TME are emerging as promising approaches for improving the efficacy of antitumor therapies by enhancing tumor perfusion and drug delivery, as well as alleviating the immunosuppressive TME. In this regard, nanomedicine has shown great potential for developing effective treatments capable of re-modeling the TME by controlling drug action in a spatiotemporal manner and allowing long-lasting modulatory effects on the TME. Herein, we review recent progress on TME re-engineering by using nanomedicine, particularly focusing on formulations controlling TME characteristics through targeted interaction with cellular components of the TME. Importantly, the TME should be re-engineering to a quiescent phenotype rather than be destroyed. Finally, immediate challenges and future perspectives of TME-re-engineering nanomedicines are discussed, anticipating further innovation in this growing field. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Takuya Miyazaki
- Kanagawa Institute of Industrial Science and Technology, Ebina, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
|
3
|
Miao T, Wang J, Zeng Y, Liu G, Chen X. Polysaccharide-Based Controlled Release Systems for Therapeutics Delivery and Tissue Engineering: From Bench to Bedside. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700513. [PMID: 29721408 PMCID: PMC5908359 DOI: 10.1002/advs.201700513] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/19/2017] [Indexed: 05/08/2023]
Abstract
Polysaccharides or polymeric carbohydrate molecules are long chains of monosaccharides that are linked by glycosidic bonds. The naturally based structural materials are widely applied in biomedical applications. This article covers four different types of polysaccharides (i.e., alginate, chitosan, hyaluronic acid, and dextran) and emphasizes their chemical modification, preparation approaches, preclinical studies, and clinical translations. Different cargo fabrication techniques are also presented in the third section. Recent progresses in preclinical applications are then discussed, including tissue engineering and treatment of diseases in both therapeutic and monitoring aspects. Finally, clinical translational studies with ongoing clinical trials are summarized and reviewed. The promise of new development in nanotechnology and polysaccharide chemistry helps clinical translation of polysaccharide-based drug delivery systems.
Collapse
Affiliation(s)
- Tianxin Miao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- School of Chemical & Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- Collaborative Innovation Center of Guangxi Biological Medicine and theMedical and Scientific Research CenterGuangxi Medical UniversityNanning530021China
| | - Yun Zeng
- Department of PharmacologyXiamen Medical CollegeXiamen361008China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamen361102China
- State Key Laboratory of Cellular Stress BiologyInnovation Center for Cell BiologySchool of Life SciencesXiamen UniversityXiamen361102China
- State Key Laboratory of Physical Chemistry of Solid Surfaces and The MOE Key Laboratory of Spectrochemical Analysis & InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamen361005China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and NanomedicineNational Institute of Biomedical Imaging and BioengineeringNational Institutes of HealthBethesdaMD20892USA
| |
Collapse
|
4
|
Basu A, Kunduru KR, Abtew E, Domb AJ. Polysaccharide-Based Conjugates for Biomedical Applications. Bioconjug Chem 2015; 26:1396-412. [DOI: 10.1021/acs.bioconjchem.5b00242] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Arijit Basu
- Institute
for Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel 91120
- Department
of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, 835215, India
| | - Konda Reddy Kunduru
- Institute
for Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel 91120
| | - Ester Abtew
- Institute
for Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel 91120
| | - Abraham J. Domb
- Institute
for Drug Research, School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel 91120
| |
Collapse
|
5
|
Goodarzi N, Varshochian R, Kamalinia G, Atyabi F, Dinarvand R. A review of polysaccharide cytotoxic drug conjugates for cancer therapy. Carbohydr Polym 2013; 92:1280-93. [DOI: 10.1016/j.carbpol.2012.10.036] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 09/27/2012] [Accepted: 10/15/2012] [Indexed: 11/30/2022]
|
6
|
Venditto VJ, Simanek EE. Cancer therapies utilizing the camptothecins: a review of the in vivo literature. Mol Pharm 2010; 7:307-49. [PMID: 20108971 DOI: 10.1021/mp900243b] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review summarizes the in vivo assessment-preliminary, preclinical, and clinical-of chemotherapeutics derived from camptothecin or a derivative. Camptothecin is a naturally occurring, pentacyclic quinoline alkaloid that possesses high cytotoxic activity in a variety of cell lines. Major limitations of the drug, including poor solubility and hydrolysis under physiological conditions, prevent full clinical utilization. Camptothecin remains at equilibrium in an active lactone form and inactive hydrolyzed carboxylate form. The active lactone binds to DNA topoisomerase I cleavage complex, believed to be the single site of activity. Binding inhibits DNA religation, resulting in apoptosis. A series of small molecule camptothecin derivatives have been developed that increase solubility, lactone stability and bioavailability to varying levels of success. A number of macromolecular agents have also been described wherein camptothecin(s) are covalently appended or noncovalently associated with the goal of improving solubility and lactone stability, while taking advantage of the tumor physiology to deliver larger doses of drug to the tumor with lower systemic toxicity. With the increasing interest in drug delivery and polymer therapeutics, additional constructs are anticipated. The goal of this review is to summarize the relevant literature for others interested in the field of camptothecin-based therapeutics, specifically in the context of biodistribution, dosing regimens, and pharmacokinetics with the desire of providing a useful source of comparative data. To this end, only constructs where in vivo data is available are reported. The review includes published reports in English through mid-2009.
Collapse
Affiliation(s)
- Vincent J Venditto
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | | |
Collapse
|
7
|
Ma H, Li X, Yang Z, Okuno S, Kawaguchi T, Yagi S, Bouvet M, Hoffman RM. High antimetastatic efficacy of MEN4901/T-0128, a novel camptothecin carboxymethyldextran conjugate. J Surg Res 2010; 171:684-90. [PMID: 20851421 DOI: 10.1016/j.jss.2010.05.066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2010] [Revised: 05/11/2010] [Accepted: 05/28/2010] [Indexed: 11/24/2022]
Abstract
The antimetastatic activity of a novel camptothecan conjugate, MEN4901/T-0128, in which 7-ethyl-10-aminopropyloxy-camptothecin (T-2513) is bound to a biodegradable carboxymethyldextran via a Gly-Gly-Gly linker, was observed in this study. High antimetastatic activity of MEN4901/T-0128 was demonstrated in a clinically-relevant orthotopic mouse model of human colon cancer. MEN4901/T-0128 and irinotecan were compared for anti-metastatic activity as well as efficacy against the primary tumor. An imageable, metastatic model was made by surgical orthotopic implantation (SOI) of the green fluorescent protein (GFP)-expressing HT-29 tumor in nude mice. MEN4901/T-0128 and irinotecan were administered intravenously at various doses and schedules. MEN4901/T-0128, with treatment beginning on d 49 after SOI, was highly effective on lymph node metastasis as well as against the primary tumor. Both GFP imaging and histology demonstrated a markedly lower metastatic incidence of lymph nodes in all MEN4901/T-0128 treated mice compared with irinotecan-treated and untreated mice. At the most efficacious dose of MEN4901/T-0128, only 1 of 12 animals had lymph node metastasis compared with 19 of 20 in the control group. The present study demonstrates the principle that when a camptothecan is conjugated to an appropriate polymer, the drug can become extremely effective with important clinical potential for antimetastatic therapy, a most urgent need.
Collapse
Affiliation(s)
- Huaiyu Ma
- AntiCancer, Inc., San Diego, California 92111, USA
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Bigioni M, Parlani M, Bressan A, Bellarosa D, Rivoltini L, Animati F, Crea A, Bugianesi R, Maggi CA, Manzini S, Binaschi M. Antitumor activity of delimotecan against human metastatic melanoma: pharmacokinetics and molecular determinants. Int J Cancer 2009; 125:2456-64. [PMID: 19536774 DOI: 10.1002/ijc.24661] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Delimotecan (MEN 4901/T-0128) is a new cytotoxic prodrug constituted by a camptothecin analog (T-2513) bound to carboxymethyl dextran through a triglycine linker. A significant antitumor activity of delimotecan against human metastatic melanoma xenograft model Me15392 is reported. Dacarbazine, the drug approved for the treatment of metastatic melanoma, was ineffective in this melanoma model. Pharmacokinetic studies, together with the expression analysis of mRNA for enzymes involved in delimotecan metabolism, showed that T-2513 and other cytotoxic metabolites of delimotecan (SN 38 and T-0055) are generated in greater quantities in the tumor tissue than in toxicity target tissues, such as liver, thus accounting for the antitumoral activity. Moreover, we demonstrated that human metastatic melanoma cells are able to phagocytose delimotecan and cleave it to release the cytotoxic moieties T-2513 in the tumoral environment. Further flow cytometric analysis showed a higher recruitment of macrophages in xenografted human metastatic melanoma, when compared with other human tumors. Thus, the antitumoral activity of delimotecan exerted on metastatic melanoma is due to several factors: (i) the ability of melanoma cells to phagocytose and metabolise delimotecan; (ii) the accumulation of delimotecan in tumoral mass; (iii) the recruitment of macrophage cells to the melanoma nodule and (iv) the expression in melanoma cells of a pattern of enzymes that converts delimotecan into cytotoxic metabolites. Based on these results, delimotecan might be exploited as a new anticancer agent for the therapy of metastatic melanoma because of its high efficacy and good selectivity, and therefore clinical trials for this indication are warranted.
Collapse
Affiliation(s)
- Mario Bigioni
- Pharmacology Department of Menarini Ricerche SpA, Pomezia, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Juan TY, Roffler SR, Hou HS, Huang SM, Chen KC, Leu YL, Prijovich ZM, Yu CP, Wu CC, Sun GH, Cha TL. Antiangiogenesis targeting tumor microenvironment synergizes glucuronide prodrug antitumor activity. Clin Cancer Res 2009; 15:4600-11. [PMID: 19584154 DOI: 10.1158/1078-0432.ccr-09-0090] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE This study is aimed at investigating the in vivo antitumor activity of a novel cell-impermeable glucuronide prodrug, 9-aminocamptothecin glucuronide (9ACG), and elucidating the synergistically antitumor effects of antiangiogenesis therapy by targeting the tumor microenvironment. EXPERIMENTAL DESIGN We analyzed the antitumor effects of 9ACG alone or combined with antiangiogenic monoclonal antibody DC101 on human tumor xenografts by measuring tumor growth and mouse survival in BALB/c nu/nu nude and NOD/SCID mice. The drug delivery, immune response, and angiogenesis status in treated tumors were assessed by high performance liquid chromatography, immunohistochemistry, and immunofluorescence assays. RESULTS We developed a nontoxic and cell-impermeable glucuronide prodrug, 9ACG, which can only be activated by extracellular beta-glucuronidase to become severely toxic. 9ACG possesses potent antitumor activity against human tumor xenografts in BALB/c nu/nu nude mice but not for tumors implanted in NOD/SCID mice deficient in macrophages and neutrophils, suggesting that these cells play an important role in activating 9ACG in the tumor microenvironment. Most importantly, antiangiogenic monoclonal antibody DC101 potentiated single-dose 9ACG antitumor activity and prolonged survival of mice bearing resistant human colon tumor xenografts by providing strong beta-glucuronidase activity and prodrug delivery through enhancing inflammatory cell infiltration and normalizing tumor vessels in the tumor microenvironment. We also show that inflammatory cells (neutrophils) were highly infiltrated in advanced human colon cancer tissues compared with normal counterparts. CONCLUSIONS Our study provides in vivo evidence that 9ACG has potential for prodrug monotherapy or in combination with antiangiognesis treatment for tumors with infiltration of macrophage or neutrophil inflammatory cells.
Collapse
Affiliation(s)
- Ting-Yi Juan
- Divisions of Urology, Graduate Institute of Life Sciences, Institute of Biomedical Sciences, Academia Sinica, National Yang-Ming University,Taipei,Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Veltkamp SA, Witteveen EO, Capriati A, Crea A, Animati F, Voogel-Fuchs M, van den Heuvel IJ, Beijnen JH, Voest EE, Schellens JH. Clinical and Pharmacologic Study of the Novel Prodrug Delimotecan (MEN 4901/T-0128) in Patients with Solid Tumors. Clin Cancer Res 2008; 14:7535-44. [DOI: 10.1158/1078-0432.ccr-08-0438] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Binaschi M, Parlani M, Bellarosa D, Bigioni M, Salvatore C, Palma C, Crea A, Maggi CA, Manzini S, Goso C. Human and murine macrophages mediate activation of MEN 4901/T-0128: a new promising camptothecin analogue-polysaccharide conjugate. Anticancer Drugs 2007; 17:1119-26. [PMID: 17075311 DOI: 10.1097/01.cad.0000236307.20339.b4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
MEN 4901/T-0128 is a new cytotoxic prodrug constituted by the camptothecin analogue T-2513 bound to carboxymethyl dextran through a triglycine linker. MEN 4901/T-0128 was designed to target the active camptothecin at the tumour site. MEN 4901/T-0128 is weakly cytotoxic in vitro and thus T-2513 must be released from the conjugate to become active. Here, we demonstrated that human purified cathepsin B releases T-2513 from MEN 4901/T-0128 at pH values ranging from 3 to 5. pH dependency of this reaction suggests that cleavage of the linker should mainly occur in the lysosomes. As elevated cathepsin B activity has been described in macrophages, human tumour monocytic THP-1 cells differentiated into macrophage-like cells were used to study the cellular mechanisms responsible for MEN 4901/T-0128 antitumour activity. Here, we show that differentiated THP-1 internalizes MEN 4901/T-0128 efficiently in a time-dependent and concentration-dependent manner. After phagocytosis, THP-1 cells can cleave the prodrug and release T-2513 in the media. On the contrary, undifferentiated THP-1 cells or pancreatic ASPC-1 tumour cells, although expressing high levels of cathepsin B, are much less efficient in the release of cytotoxic moieties in the culture media. Moreover, normal murine macrophages, recovered from the peritoneal cavity or from the spleen, when activated (in vitro by 100 ng/ml phorbol 12-myristate-13-acetate and in vivo by 300 microl of 3% w/v thioglycollate solution), were able to release (after incubation with 10 microg/ml MEN 4901/T-0128) cytotoxic moieties in the culture supernatant, in an amount sufficient to kill human carcinoma A2780 cells. Thus, we suggest that tumour-associated macrophages may play a key role in the uptake of MEN 4901/T-0128, cleavage and local release of active moiety T-2513. This mechanism should support a tumour targeting of the cytotoxic moieties, allowing an improved antitumour efficacy/safety ratio for MEN 4901/T-0128.
Collapse
|
12
|
Sugahara SI, Kajiki M, Kuriyama H, Kobayashi TR. Complete regression of xenografted human carcinomas by a paclitaxel-carboxymethyl dextran conjugate (AZ10992). J Control Release 2006; 117:40-50. [PMID: 17126446 DOI: 10.1016/j.jconrel.2006.10.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 10/05/2006] [Accepted: 10/08/2006] [Indexed: 11/30/2022]
Abstract
Clinically available taxanes, such as paclitaxel and docetaxel, represent one of the most promising classes of anticancer agents, despite their toxicity. To improve their pharmacological profiles, AZ10992 was synthesized based on the concept that a rational design of a polymer-drug conjugate would increase the efficacy of the parent drug. This prodrug is a paclitaxel-carboxymethyl dextran conjugate (molecular weight 150,000 g/mol) via a gly-gly-phe-gly linker. The in vivo antitumor study using AZ10992 against colon26 carcinoma cells, resistant to paclitaxel, supported this concept. Additionally, the comparative efficacy studies of AZ10992 and paclitaxel using a panel of human tumor xenografts in nude mice showed the advantages of drug-polymer conjugation. The maximum tolerated dose of AZ10992 was more than twice as high as the MTD of paclitaxel. A repeated intravenous administration of AZ10992 at 30 mg/kg/day (five injections for 4-days) showed complete regression of MX-1 mammary carcinoma xenografts. Also, HT-29 colorectal tumor xenografts, which are highly refractory to paclitaxel, showed complete regression after AZ10992 administered at 30 mg/kg/day (seven injections for 4-days). Pharmacokinetic studies showed that there were significant increases in the amount and the exposure time of total paclitaxel in the tumors after intravenous administration of AZ10992, which explains the enhanced efficacy of AZ10992.
Collapse
Affiliation(s)
- Shu-Ichi Sugahara
- The Second Research Department of Central Technology Laboratory, Asahi Kasei Corporation, 2-1 Samejima, Fuji, Shizuoka 416-8501, Japan.
| | | | | | | |
Collapse
|
13
|
Yoshizawa H, Nishino S, Shiomori K, Natsugoe S, Aiko T, Kitamura Y. Surface morphology control of polylactide microspheres enclosing irinotecan hydrochloride. Int J Pharm 2005; 296:112-6. [PMID: 15885462 DOI: 10.1016/j.ijpharm.2005.02.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2004] [Revised: 01/17/2005] [Accepted: 02/06/2005] [Indexed: 11/23/2022]
Abstract
In order to reduce the initial burst from polylactide (PLA) microspheres enclosing an antitumor agent, we prepared the microspheres with a smooth surface by varying solvent evaporation conditions such as operating temperature and pressure. PLA microspheres enclosing irinotecan hydrochloride (CPT) were prepared using the O/O emulsion system for solvent evaporation. The mean diameter and enclosing efficiency were almost constant because they were independent of solvent evaporation conditions. Scanning electron microscopic (SEM) observation verified the smooth surface of the PLA microspheres produced by varying the preparation conditions. In vitro release experiments show that the initial burst of microspheres with a smooth surface was less than that of those with a rough surface.
Collapse
Affiliation(s)
- Hidekazu Yoshizawa
- Department of Environmental Chemistry and Materials, Faculty of Environmental Science and Technology, Okayama University, Okayama 700-8530, Japan.
| | | | | | | | | | | |
Collapse
|
14
|
Ochi Y, Shiose Y, Kuga H, Kumazawa E. A possible mechanism for the long-lasting antitumor effect of the macromolecular conjugate DE-310: mediation by cellular uptake and drug release of its active camptothecin analog DX-8951. Cancer Chemother Pharmacol 2004; 55:323-332. [PMID: 15517271 DOI: 10.1007/s00280-004-0911-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2004] [Accepted: 08/25/2004] [Indexed: 11/29/2022]
Abstract
DE-310, a new macromolecular prodrug, was designed to enhance the pharmacological profiles of a novel camptothecin analog (DX-8951f), and a single treatment with DE-310 exhibits a similar or greater therapeutic effect than do optimally scheduled multiple administrations of DX-8951f in several types of tumors. In this study, the drug-release mechanism by which DE-310 excites antitumor activity was investigated in Meth A cells, a malignant ascites model of murine fibrosarcoma. A single i.v. injection of DE-310 at the maximum tolerated dose (MTD) prolonged survival of Meth A-bearing mice by 300%. DX-8951 and glycyl-8951 (G-DX-8951), enzymatic cleavage products of DE-310, were detected in serum and ascites fluid, and also in the culture medium of Meth A ascites cells incubated in vitro with DE-310. The total amounts of DX-8951, G-DX-8951, and conjugated DX-8951 in Meth A tumor cells were three times higher than that in macrophages. Furthermore, DX-8951-related fluorescence was observed in Meth A ascites cells obtained from Meth A-bearing mice that had received DE-310 or CM-Dex-PA-DX-8951 that does not release free DX-8951. DX-8951-related fluorescence was also observed at the site of lysosomes in cells incubated in vitro with DE-310 at 37 degrees C, but not in those incubated at 4 degrees C. Drugs were released from DE-310 by cysteine proteinase prepared from Meth A tumor tissue. These results suggest that the mechanism by which DX-8951 is released from DE-310 in vivo is involved in the process of uptake of DE-310 into tumor or macrophages, digestion by intracellular lysosomal cysteine proteinase, and subsequent secretion of the drugs.
Collapse
Affiliation(s)
- Yusuke Ochi
- New Product Research Laboratories III, Daiichi Pharmaceutical Co., Ltd., Tokyo R&D Center, 16-13, Kita-Kasai 1-Chome, Edogawa-ku, Tokyo, 134-8630, Japan.
| | - Yoshinobu Shiose
- New Product Research Laboratories III, Daiichi Pharmaceutical Co., Ltd., Tokyo R&D Center, 16-13, Kita-Kasai 1-Chome, Edogawa-ku, Tokyo, 134-8630, Japan
| | - Hiroshi Kuga
- New Product Research Laboratories III, Daiichi Pharmaceutical Co., Ltd., Tokyo R&D Center, 16-13, Kita-Kasai 1-Chome, Edogawa-ku, Tokyo, 134-8630, Japan
| | - Eiji Kumazawa
- New Product Research Laboratories III, Daiichi Pharmaceutical Co., Ltd., Tokyo R&D Center, 16-13, Kita-Kasai 1-Chome, Edogawa-ku, Tokyo, 134-8630, Japan
| |
Collapse
|
15
|
Yoshizawa H, Nishino S, Natsugoe S, Aiko T, Kitamura Y. Surface Morphology Change of Polylactide Microspheres Enclosing Irinotecan Hydrochloride and Its Effect on Release Properties. JOURNAL OF CHEMICAL ENGINEERING OF JAPAN 2003. [DOI: 10.1252/jcej.36.1206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Hidekazu Yoshizawa
- Department of Environmental Chemistry and Materials, Faculty of Environmental Science and Technology, Okayama University
| | - Satoru Nishino
- Department of Environmental Chemistry and Materials, Faculty of Environmental Science and Technology, Okayama University
| | - Shoji Natsugoe
- First Department of Surgery, Kagoshima University, School of Medicine
| | - Takashi Aiko
- First Department of Surgery, Kagoshima University, School of Medicine
| | - Yoshiro Kitamura
- Department of Environmental Chemistry and Materials, Faculty of Environmental Science and Technology, Okayama University
| |
Collapse
|
16
|
Harada M, Murata J, Sakamura Y, Sakakibara H, Okuno S, Suzuki T. Carrier and dose effects on the pharmacokinetics of T-0128, a camptothecin analogue-carboxymethyl dextran conjugate, in non-tumor- and tumor-bearing rats. J Control Release 2001; 71:71-86. [PMID: 11245909 DOI: 10.1016/s0168-3659(00)00372-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
T-0128 is a novel camptothecin (CPT) analogue (T-2513: 7-ethyl-10-aminopropyloxy-CPT)-carboxymethyl (CM) dextran conjugate via a Gly-Gly-Gly linker, with a molecular weight (MW) of 130 kDa. Our previous studies demonstrated that T-0128 has strong antitumor activity against human tumor xenografts that are highly refractory to CPT analogues attributable to the passive tumor targeting of released T-2513. This study examines the effects of carrier, dose, and tumor on T-0128 pharmacokinetics. To study carrier effect, tumor-bearing rats received one i.v. injection of fluorescein isothiocyanate (FITC)-labeled CM dextran with a different degree of substitution (DS) of the carboxymethylated groups and a different MW. Results showed that CM dextran from Dextran T-110 (MW 110 kDa) with a DS value of 0.4 is an appropriate drug carrier for T-0128 regarding plasma half-life and passive tumor targeting. To study dose and tumor effects, non-tumor- and tumor-bearing rats were treated with T-0128 doses ranging from 1 to 25 mg/kg (based on the amount of T-2513 bound to CM dextran). Dose-dependent pharmacokinetics of T-0128 were observed in both kinds of rats. The presence of tumor reduced the plasma half-life and systemic exposure of T-0128. The saturation of hepatic and splenic tissue uptake clearances (CLups), and a large contribution of the tumor CLup to the total body clearance explain these results. Overall, our data provide a rationale for the selection of the carrier for T-0128 and a need for pharmacokinetic studies to evaluate the influences of tumor on the drug disposition.
Collapse
Affiliation(s)
- M Harada
- Discovery Research Laboratory, Tanabe Seiyaku Co., Ltd., Kashima 3-16-89, Yodogawa-ku, 532-8505, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
17
|
Harada M, Sakakibara H, Yano T, Suzuki T, Okuno S. Determinants for the drug release from T-0128, camptothecin analogue-carboxymethyl dextran conjugate. J Control Release 2000; 69:399-412. [PMID: 11102680 DOI: 10.1016/s0168-3659(00)00321-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
To improve pharmacological profiles of camptothecins (CPTs), a new macromolecular prodrug, denoted T-0128, was synthesized. This prodrug comprises a novel CPT analog (T-2513: 7-ethyl-10-aminopropyloxy-CPT) bound to carboxymethyl (CM) dextran through a Gly-Gly-Gly linker, with a molecular weight of 130 kDa. The present study was designed to elucidate the mechanisms that promote the release of linked T-2513. First, we compared the abilities of a rat liver homogenate, a cocktail of its lysosomal enzymes, and different types of pure enzymes, to liberate T-2513 from the conjugate. The releasing rate in the homogenate was very slow, but was accelerated with the lysosomes. Lysosomal cysteine proteinases, such as cathepsin B, were responsible, coupled with the results of in vitro and in vivo inhibition studies using proteinase inhibitors. The pH optimum for the cathepsin B-mediated drug release was approximately 4. This corresponds to the pH in lysosomes, suggesting lysosomotropic release. Second, to assess the effect of the length and composition of the peptidyl linker, we synthesized the conjugates with a different linker and compared the drug-releasing rates. We found that the insertion of Phe into Gly-Gly-Gly allowed various kinds of enzymes to produce a rapid cleavage, and the Gly-chain lengthening enhanced the lysosome-mediated drug release. The released T-2513 levels in the liver and tumor of the tumor-bearing rats dosed with each conjugate increased with the length of Gly linker, suggesting a good in vitro to in vivo relationship. Comparative efficacy studies of the conjugates with a different linker demonstrated that T-0128 showed the maximum efficacy against MX-1 human mammary xenograft tumors. Thus the Gly-Gly-Gly linker exploits lysosomal cathepsin B to liberate T-2513 slowly and steadily, resulting in improved therapeutic efficacy.
Collapse
Affiliation(s)
- M Harada
- Discovery Research Laboratory, Tanabe Seiyaku Co. Ltd., Kashima 3-16-89, Yodogawa-ku, 532-8505, Osaka, Japan.
| | | | | | | | | |
Collapse
|