Publications

Detailed Information

Designing functional formulations of a taurocholate conjugated low molecular weight heparin derivative for targeting tumor angiogenesis and metastasis

DC Field Value Language
dc.contributor.advisor이동수-
dc.contributor.author파자나-
dc.date.accessioned2017-07-14T01:51:15Z-
dc.date.available2017-07-14T01:51:15Z-
dc.date.issued2015-02-
dc.identifier.other000000025028-
dc.identifier.urihttps://hdl.handle.net/10371/122403-
dc.description학위논문 (박사)-- 서울대학교 융합과학기술대학원 : 분자의학 및 바이오제약학과, 2015. 2. 이동수.-
dc.description.abstractHeparin is a biomimetic molecule, which is commercially used as an anti-coagulant drug. But years of research have shown its efficacy in many other biological systems, such as, in immunology, angiogenesis and metastasis. Among all of the above mentioned effects, the anti-cancer effect of heparin showed promising results in many studies. However, unfractionated heparin cannot be used as anti-cancer drug because of its side effects, such as- heparin induced thrombocytopenia (HIT). So, low molecular weight heparin (LMWH) and its chemically modified heparin derivatives are under extensive studies as anti-cancer drugs in many cancer related fields. Previously, we synthesized several low molecular weight heparin moieties, which can be used as anticancer drugs. Among them seven taurocholate conjugated low molecular weight heparin derivative (LHT7) showed significant effect as an angiogenesis inhibitor. In our previous studies, LHT7 proved as highly effective in the inhibition of tumor angiogenesis by targeting multiple growth factors, such as, VEGF, bFGF, PDGF, HGF, etc. However, during safety evaluation in preclinical study, LHT7 showed unwanted accumulation in liver and kidney and thus showed functional abnormalities of these organs. Moreover, the half-life of LHT7 is short (2 h) and thus the modification in the delivery through either the change in administration or the development of effective formulation is necessary to increase the half-life with lowering the toxicological effect of this noble molecule. Finally, intravenous (i.v) administration of LHT7 could be less desirable to some patients for the maintenance anti-cancer therapy. So, developing patient compatible oral formulation is also a necessary part of the LHT7s development.
As the LHT7 showed toxic effects upon daily administration to the animals in preclinical studies, we at first change the administration condition from i.v blous form to i.v infusion. At first, for the evaluation of safety, the method of intravenous infusion was compared with that of i.v bolus (at rate 400 μl/min/kg of body weight for 30 min). Then, for pharmacokinetic analysis, organ accumulation and plasma concentration profiles of LHT7 were measured. Furthermore, the anticancer effects of LHT7 were measured in murine (SCC7) and human xenograft (MDAMB-231) model, and the general safety studies were performed in beagle dogs. The result of the pharmacokinetic studies showed that the reduced organ accumulation in mice and the AUC(0-96h) was increased up to 1257.38 ± 0.11 h*μg/m. The efficacy, at dose 1mg/kg/2 days was higher for i.v infusion than for i.v bolus administration in both murine and human cancer model. The preliminary safety analysis of SD rats showed that there were no organ specific side effects in higher doses. So, from our study, we concluded that, LHT7 showed sustained effects with minimized adverse events when it is administered through i.v infusion. It was also shown that a maximum dose of 12 mg/kg (through i.v infusion) could be safely used for further clinical development of LHT7 as a multi-targeting anti-angiogenic agent.
On the other hand, drugs that have been designed to block angiogenesis mainly capture growth factors in circulation, resulting not only in the transient inhibition of tumor progression but also in producing undesirable side effects. Nanoparticular drug delivery systems, on the other hand, may help overcome such drawbacks and improve the efficacy of anti-angiogenic therapies by altering the biodistribution and pharmacokinetics, improving tumor targeting ability, and reducing side effects. In this light, we propose a new approach of anti-angiogenic therapy that combines strategies of long circulating, passive tumor targeting, and anti-angiogenesis efficacy using a new polyelectrolyte complex system that combines LHT7, with a protamine to form a self-assembling nanocomplex with a mean diameter of 200 nm with zeta potential of -14.2 ± 1.2 mV, which is designed to produce effective anti-angiogenic effect. At first, LHT7 was modified with poly-ethylene-glycol (PEG) and form nanocomplex with positively charged protamine (in buffer). We observed that PEG-LHT7/protamine nanocomplex was stable in buffer and slowly dissociated in plasma (9% dissociation after 24 h). Compared to the free form of PEG-LHT7, the mean residence time of PEG-LHT7/protamine nanocomplex was found higher (15.9 h) with its increased accumulation in tumor that was found in biodistribution study. Most importantly, PEG-LHT7/protamine nanocomplex was diffused and extravasated through the dense collagen matrix of tumor. Although the nanocomplex showed accumulation in liver, beside tumor in the biodistribution study, the serological evaluation of this nanocomplex did not show any functional variation of liver. Thus, the study describes a successful application of functionalized PEG-LHT/protamine nanocomplex that can inhibit angiogenesis with long circulating, passive targeting, and tumor extravasating ability with minimized side effect.
Taurocholate conjugated low molecular weight heparin derivative (LHT7) has been proven to be a potent, multi-targeting angiogenesis inhibitor against broad-spectrum angiogenic tumors. However, major limitations of LHT7 are its poor oral bioavailability, short half-life, and frequent parenteral dosing schedule. Addressing these issues, we have developed an oral formulation of LHT7 by chemically conjugating LHT7 with a tetrameric deoxycholic acid named LHTD4, and then physically complexing it with deoxycholylethylamine (DCK). The resulting LHTD4/DCK complex showed significantly enhanced oral bioavailability (34.3 ± 2.89%) and prolonged the mean residence time (7.5 ± 0.5 h). The LHTD4/DCK complex was mostly absorbed in the intestine by transcellular pathway via its interaction with apical sodium bile acid transporter. Moreover, in biodistribution study, LHT7 mainly showed increased accumulationin tumor after 30 min of administration. In vitro, the VEGF-induced sprouting of endothelial spheroids was significantly blocked by LHTD4. LHTD4/DCK complex significantly regressed the total vessel fractions of tumor (77.2 ± 3.9%), as analyzed by X-ray microCT angiography, thereby inhibiting tumor growth in vivo. Using the oral route of administration, we showed that LHTD4/DCK complex could be effective and chronically administered as angiogenesis inhibitor.
Targeting multiple stages in metastatic breast cancer is one of the effective ways to inhibit metastatic progression. As metastasis is a longer process, so the therapy should be more compatible to the patients. Considering these theories, we used an orally active polymeric bile acetylated taurocholate conjugated low molecular weight heparin derivative (LHTD4) with a formulation containing synthetic bile acid enhancer (DCK) to target human metastasis breast cancer. In case of breast cancer, TGFβ1 and CXCL12 possess enhanced metastatic activity during the initiation and the progression (seeding in other organs). So, in our study, we focus the binding effect of LHTD4 with TGFβ1 and CXCL12. We carried out computer simulation study and SPR analysis for the binding confirmation of LHTD4 with TGFβ1 and CXCL12. Here, we found that the KD values of TGFβ1 and CXCL12 with LHTD4 were 0.85 and 0.019 µM respectively. Moreover, the simulation showed stable binding affinities of the dp4 moieties of LHTD4 with through the strong electrostatic interaction. After confirming the binding affinity, we carried out in vitro phosphorylation assays of the consecutive receptors of TGFβ1 and CXCL12 (TGFβ1R1 and CXCR4 receptor respectively)
-
dc.description.abstractwhere the successful inhibition of the phosphorylation of those receptors was observed with the treatment of LHTD4. The expression changes of EMT marker proteins, such as, E-cadherin (degradation), Vimentin (increased expression) and SNAIL (increased expression), were prevented by the LTHD4 treatment in our in vitro studies with TGFβ1 treated MDAMB231 cells. Moreover, cell migration (induced by TGFβ1) and chemotaxis (mediated by CXCL12) of MDAMB-231 cells were inhibited by LHTD4 treatment. Finally, through accelerated lung metastasis model and by orthotopic MDAMB-231 breast cancer model, the metastasis inhibitory effect was evaluated in the mice. Finally, through metastasis inhibition analysis of the breast cancer cells in in vivo confirmed the anti-metastatic effect of LHTD4/DCK by inhibiting TGFβ1 and CXCL12.
In conclusion, LHT7 is a noble molecule, which can be applied clinically to target tumor angiogenesis as a second line cancer therapy. With i.v infusion administration, LHT7 can be effective and thus increased tumor accumulation can showed less organ accumulation and reduced toxic events. More over, nanocomplex of PEGylated LHT7 with protamine showed increased the circulation time and tumor accumulation
-
dc.description.abstractthus proved to be reasonable to apply to the patients to avoid daily administration. Finally, for patient compliance and maintenance therapy oral formulation showed more applicability in the outdoor therapy with similar efficacy. Moreover, besides targeting angiogenesis, oral tetra-deoxycholate conjugated LHT7, LHTD4 can effectively use as an anti-metastasis drug by targeting TGFβ1 and CXCL12, which play key roles in the initiation and development of breast cancer metastasis.-
dc.description.tableofcontentsAbstract i
Table of contents vi
Lists of Tables xi
Lists of Figures xii
Abbreviations xv
Chapter 1. Introduction of heparin derivatives as anti-cancer drug
1.1 Introduction 2
1.2 Heparin and low molecular weight heparin: Structure activity relationship 3
1.3 The effect of heparin on cancer 4
1.4 Taurocholate conjugated low molecular weight heparin derivative 7
1.4.1 Chemistry, targeting and efficiency 7
1.4.2 Preclinical and general safety study 10
1.5 Rationale of the study 11
1.6 Reference 16
Chapter 2. Functionalized self-assembled bile-acetylated heparin-protamine nanocomplex for targeting angiogenesis
2.1 Introduction 20
2.1.1 Advantages of nanoparticles in anti-cancer drug delivery systems 20
2.1.2 Introduction to Poly electric complex system 21
2.1.3 Purpose of the study 22
2.2 Materials and methods 23
2.2.1 Materials 23
2.2.2 Synthesis of PEG-LHT7 24
2.2.3 Formulation of nanocomplex 24
2.2.4 Characterization of nanaocomplex 26
2.2.5 Pharmacokinetic profile study 26
2.2.6 Ex-vivo tumor accumulation and distribution study 28
2.2.7 In-vivo anti-angiogenic effect 29
2.2.8 X-ray μCT angiography 30
2.2.9 Toxicological analysis 31
2.3 Results 31
2.3.1 Chemical characterization of PEG-LHT7/protamine nanocomplex 31
2.3.2 In-vitro release study 33
2.3.3 Biodistribution of PEG-LHT7/protamine nanocomplex 35
2.3.4 Pharmacokinetics of PEG-LHT7/protamine nanocomplex 35
2.3.5 Intratumoral localization of PEG-LHT7/protamine nanocomplex 39
2.3.6 Anticancer effect of PEG-LHT7/protamine nanocomplex by the regression of angiogenesis 39
2.3.7 Preliminary toxicity study 49
2.4 Discussion 49
2.5 Conclusion 54
2.6 Reference 55
Chapter 3. Formulation of an orally active physical complex of tetra-DOCA acetylated taurocholate conjugated low molecular heparin derivative as an anti-angiogenic agent with synthetic bile acid enhancer
3.1 Introduction 61
3.1.1 Introduction of oral delivery 61
3.1.2 Role of angiogenesis in cancer 62
3.1.3 Importance of angiogenesis targeting with oral delivery 62
3.1.4 Low molecular weight as angiogenesis inhibitor 63
3.1.5 Development of oral formulation of LHT7 64
3.2 Materials and methods 66
3.2.1 Synthesis and characterization of LHTD4 66
3.2.2 Preparation of LHTD4/DCK formulation 68
3.2.3 In-vitro permeability assay 70
3.2.4 In-vivo absorption study 70
3.2.5 In-vitro anti-angiogenic effect of LHTD4 by VEGF-induced sprouting assay 72
3.2.7 Inhibition of tumor growth in murine SCC7 model 72
3.2.8 X-ray μCT angiography 73
3.2.9 Statistical analysis 74
3.3 Results 74
3.3.1 Characterizations of LHTD4 and its physical complex with DCK 74
3.3.2 Inhibition effect of LHTD4 on sprouting of EC spheroids 74
3.3.3 Absorption mechanism of LHTD4 and its DCK complex in the intestine 75
3.3.4 Pharmacokinetic study 77
3.3.5 Inhibition effect of oral LHTD4/DCK complex on tumor growth 77
3.4 Discussion 80
3.5 Conclusion 85
3.6 Reference 86
Chapter 4. Targeting multiple stages of metastasis using orally active bile acetylated low molecular weight heparin derivative (LHTD4)
4.1 Introduction 91
4.1.1 Role of metastasis in the cancer progression and therapy 91
4.1.2 Importance of metastasis and anti-metastatic targets for the breast cancer treatment 93
4.1.3 Role of TGFβ1 and CXCL12 in metastasis 95
4.1.4 Anti-metastatic effect oh heparin 96
4.1.5 Objective of the study 98
4.2 Materials and Methods 99
4.2.1 Material 99
4.2.2 Binding affinity and simulation study with LHTD4 with TGFβ1 and CXCL12 100
4.2.3 Cell culture 101
4.2.4 Cell migration and chemotaxsis assay induced by TGFβ1 and CXCL12 respectively 101
4.2.5 Role of LHTD4 in epithelial to mesenchymal transition (EMT) related protein expression induced by TGFβ1 102
4.2.6 In vitro evaluation of TGFβ1R1 receptor phosphorylation by using LHTD4 103
4.2.7 In vitro evaluation of CXCR4 receptor phosphorylation by using LHTD4 104
4.2.8 In vivo anti-metastatic effect of LHTD4 in murine breast cancer cell line 4T1-LUC-tdmt orthotopic and resection model 105
4.2.9 In vivo anti-metastatic-growth in accelerated metastatic model using human breast cancer cell line MDA-MB-231-LUC2 cell line 106
4.2.10 In vivo anti-metastatic-spread effect of LHTD4/DCK in orthotopic model using MDA-MB-231-LUC2 cell line 106
4.3 Results 107
4.3.1 Chemistry and binding affinity of LHTD4 with TGFβ1 and CXCL12 107
4.3.2 Effect of LMWH and LHTD4 on TGFβ1-mediated signaling and its regulation on EMT in breast cancer cell line 111
4.3.3 Effect of LMWH and LHTD4 on CXCL12-Mediated signaling and its regulation on breast cancer cell invasion 112
4.3.4 LHTD4 Inhibits metastases of murine orthotopic mammary Tumor 119
4.3.5 In vivo metastasis inhibition of breast cancer models with the treatment of LHTD4/DCK 120
4.3.6 Preventive effects of LHTD4/DCK on experimental lung metastasis of human breast cancer in-vivo 127
4.4 Discussion 128
4.5 Conclusion 130
4.6 Reference 132
Chapter 5. Concluding Remarks 137
-
dc.formatapplication/pdf-
dc.format.extent6467923 bytes-
dc.format.mediumapplication/pdf-
dc.language.isoen-
dc.publisher서울대학교 대학원-
dc.subjectTaurocholate conjugated low molecular weight heparin (LHT7)-
dc.subjectinfusion-
dc.subjectangiogenesis-
dc.subjectnanocomplex-
dc.subjectmeatstasis-
dc.subjectoral delivery-
dc.subject.ddc610-
dc.titleDesigning functional formulations of a taurocholate conjugated low molecular weight heparin derivative for targeting tumor angiogenesis and metastasis-
dc.typeThesis-
dc.contributor.AlternativeAuthorFarzana Alam-
dc.description.degreeDoctor-
dc.citation.pagesxvii, 148-
dc.contributor.affiliation융합과학기술대학원 분자의학 및 바이오제약학과-
dc.date.awarded2015-02-
Appears in Collections:
Files in This Item:

Altmetrics

Item View & Download Count

  • mendeley

Items in S-Space are protected by copyright, with all rights reserved, unless otherwise indicated.

Share