Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
NPJ Precis Oncol ; 8(1): 94, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654141

ABSTRACT

Trophoblast cell surface antigen 2 (Trop2) is considered to be an attractive therapeutic target in cancer treatments. We previously generated a new humanized anti-Trop2 antibody named hIMB1636, and designated it as an ideal targeting carrier for cancer therapy. Lidamycin (LDM) is a new antitumor antibiotic, containing an active enediyne chromophore (AE) and a noncovalently bound apoprotein (LDP). AE and LDP can be separated and reassembled, and the reassembled LDM possesses cytotoxicity similar to that of native LDM; this has made LDM attractive in the preparation of gene-engineering drugs. We herein firstly prepared a new fusion protein hIMB1636-LDP composed of hIMB1636 and LDP by genetic engineering. This construct showed potent binding activities to recombinant antigen with a KD value of 4.57 nM, exhibited binding to Trop2-positive cancer cells and internalization and transport to lysosomes, and demonstrated powerful tumor-targeting ability in vivo. We then obtained the antibody-drug conjugate (ADC) hIMB1636-LDP-AE by molecular reconstitution. In vitro, hIMB1636-LDP-AE inhibited the proliferation, migration, and tumorsphere formation of tumor cells with half-maximal inhibitory concentration (IC50) values at the sub-nanomolar level. Mechanistically, hIMB1636-LDP-AE induced apoptosis and cell-cycle arrest. In vivo, hIMB1636-LDP-AE also inhibited the growth of breast and lung cancers in xenograft models. Moreover, compared to sacituzumab govitecan, hIMB1636-LDP-AE showed more potent antitumor activity and significantly lower myelotoxicity in tumors with moderate Trop2 expression. This study fully revealed the potent antitumor efficacy of hIMB1636-LDP-AE, and also provided a new preparation method for LDM-based ADC, as well as a promising candidate for breast cancer and lung cancer therapeutics.

2.
J Med Chem ; 66(21): 14700-14715, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37883180

ABSTRACT

Herein, we first prepared a novel anti-TROP2 antibody-drug conjugate (ADC) hIMB1636-MMAE using hIMB1636 antibody chemically coupled to monomethyl auristatin E (MMAE) via a Valine-Citrulline linker and then reported its characteristics and antitumor activity. With a DAR of 3.92, it binds specifically to both recombinant antigen (KD ∼ 0.687 nM) and cancer cells and could be internalized by target cells and selectively kill them with IC50 values at nanomolar/subnanomolar levels by inducing apoptosis and G2/M phase arrest. hIMB1636-MMAE also inhibited cell migration, induced ADCC effects, and had bystander effects. It displayed significant tumor-targeting ability and excellent tumor-suppressive effects in vivo, resulting in 5/8 tumor elimination at 12 mg/kg in the T3M4 xenograft model or complete tumor disappearance at 10 mg/kg in BxPc-3 xenografts in nude mice. Its half-life in mice was about 87 h. These data suggested that hIMB1636-MMAE was a promising candidate for the treatment of pancreatic cancer with TROP2 overexpression.


Subject(s)
Immunoconjugates , Pancreatic Neoplasms , Humans , Animals , Mice , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Cell Line, Tumor , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
3.
Int J Biol Macromol ; 253(Pt 6): 127105, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37769779

ABSTRACT

Trophoblast cell surface antigen 2 (Trop2) has emerged as a potential target for effective cancer therapy. In this study, we report a novel anti-Trop2 antibody IMB1636, developed using hybridoma technology. It exhibited high affinity and specificity (KD = 0.483 nM) in binding both antigens and cancer cells, as well as human tumor tissues. hIMB1636 could induce endocytosis, and enabled targeted delivery to the tumor site with an in vivo retention time of 264 h. The humanized antibody hIMB1636, acquired using CDR grafting, exhibited the potential to directly inhibit cancer cell proliferation and migration, and to induce ADCC effects. Moreover, hIMB1636 significantly inhibited the growth of MDA-MB-468 xenograft tumors in vivo. Mechanistically, hIMB1636 induced cell cycle arrest and apoptosis by regulating cyclin-related proteins and the caspase cascade. In comparison to commercialized sacituzumab, hIMB1636 recognized a conformational epitope instead of a linear one, bound to antigen and cancer cells with similar binding affinity, induced significantly more potent ADCC effects against cancer cells, and displayed superior antitumor activities both in vitro and in vivo. The data presented in this study highlights the potential of hIMB1636 as a carrier for the formulation of antibody-based conjugates, or as a promising candidate for anticancer therapy.


Subject(s)
Immunoconjugates , Neoplasms , Humans , Cell Adhesion Molecules , Antibodies, Monoclonal , Neoplasms/drug therapy , Immunoconjugates/pharmacology , Cell Proliferation , Cell Line, Tumor , Xenograft Model Antitumor Assays
4.
J Pharm Anal ; 12(2): 232-242, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35582405

ABSTRACT

Folate receptor (FR) overexpression occurs in a variety of cancers, including pancreatic cancer. In addition, enhanced macropinocytosis exists in K-Ras mutant pancreatic cancer. Furthermore, the occurrence of intensive desmoplasia causes a hypoxic microenvironment in pancreatic cancer. In this study, a novel FR-directed, macropinocytosis-enhanced, and highly cytotoxic bioconjugate folate (F)-human serum albumin (HSA)-apoprotein of lidamycin (LDP)-active enediyne (AE) derived from lidamycin was designed and prepared. F-HSA-LDP-AE consisted of four moieties: F, HSA, LDP, and AE. F-HSA-LDP presented high binding efficiency with the FR and pancreatic cancer cells. Its uptake in wild-type cells was more extensive than in K-Ras mutant-type cells. By in vivo optical imaging, F-HSA-LDP displayed prominent tumor-specific biodistribution in pancreatic cancer xenograft-bearing mice, showing clear and lasting tumor localization for 360 h. In the MTT assay, F-HSA-LDP-AE demonstrated potent cytotoxicity in three types of pancreatic cancer cell lines. It also induced apoptosis and caused G2/M cell cycle arrest. F-HSA-LDP-AE markedly suppressed the tumor growth of AsPc-1 pancreatic cancer xenografts in athymic mice. At well-tolerated doses of 0.5 and 1 mg/kg, (i.v., twice), the inhibition rates were 91.2% and 94.8%, respectively (P<0.01). The results of this study indicate that the F-HSA-LDP multi-functional bioconjugate might be effective for treating K-Ras mutant pancreatic cancer.

5.
Biochem Pharmacol ; 201: 115057, 2022 07.
Article in English | MEDLINE | ID: mdl-35489393

ABSTRACT

KRAS mutation and NF-κB both play crucial role in pancreatic cancer; in addition, defensin, the peptide mediator in innate immunity, can inhibit NF-κB. Assuming a strategy that targets both NF-κB and concomitantly the mutated KRAS indirectly via intensive macropinocytosis, we designed and generated a recombinant protein DF2-HSA which consists of two molecules of human beta-defensin 2 (HBD2) and a moiety of human serum albumin (HSA). As shown, the recombinant protein DF2-HSA markedly down-regulated NF-κB in both KRAS mutant MIA PaCa-2 cells and wild type BxPC-3 cells. Determined by confocal microscopy, the uptake of DF2-HSA in MIA PaCa-2 cells was more intense than that in BxPC-3 cells. The uptake was blocked by the specific inhibitor EIPA, indicating that DF2-HSA internalized via macropinocytosis. DF2-HSA displayed more potent cytotoxicity to cancer cells than HBD2. DF2-HSA induced apoptosis in cancer cells. Notably, DF2-HSA inhibited tumor cell spheroid formation, an effect comparable to that of salinomycin. DF2-HSA inhibited tumor cell migration and invasion. As detected with scanning electron microscopy, DF2-HSA strongly depleted filopodia on cell surface; and salinomycin induced similar changes. By in vivo imaging, DF2-HSA displayed intense tumor-site accumulation and lasting retention for over 14 days; however, HBD2 showed much less tumor-site accumulation and a shorter retention time for only 24 h. DF2-HSA suppressed the growth of pancreatic cancer MIA PaCa-2 xenograft in athymic mice; and its combination with gemcitabine achieved higher antitumor efficacy. In summary, the recombinant defensin/HSA fusion protein that inhibits NF-κb associated with intensive macropinocytosis is highly effective against pancreatic cancer.


Subject(s)
NF-kappa B , Pancreatic Neoplasms , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Mice , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Serum Albumin, Human/metabolism , Serum Albumin, Human/pharmacology , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
6.
Mil Med Res ; 8(1): 63, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34879870

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is the most aggressive subtype and occurs in approximately 15-20% of diagnosed breast cancers. TNBC is characterized by its highly metastatic and recurrent features, as well as a lack of specific targets and targeted therapeutics. Epidermal growth factor receptor (EGFR) is highly expressed in a variety of tumors, especially in TNBC. LR004-VC-MMAE is a new EGFR-targeting antibody-drug conjugate produced by our laboratory. This study aimed to evaluate its antitumor activities against EGFR-positive TNBC and further studied its possible mechanism of antitumor action. METHODS: LR004-VC-MMAE was prepared by coupling a cytotoxic payload (MMAE) to an anti-EGFR antibody (LR004) via a linker, and the drug-to-antibody ratio (DAR) was analyzed by HIC-HPLC. The gene expression of EGFR in a series of breast cancer cell lines was assessed using a publicly available microarray dataset (GSE41313) and Western blotting. MDA-MB-468 and MDA-MB-231 cells were treated with LR004-VC-MMAE (0, 0.0066, 0.066, 0.66, 6.6 nmol/L), and the inhibitory effects of LR004-VC-MMAE on cell proliferation were examined by CCK-8 and colony formation. The migration and invasion capacity of MDA-MB-468 and MDA-MB-231 cells were tested at different LR004-VC-MMAE concentrations (2.5 and 5 nmol/L) with wound healing and Transwell invasion assays. Flow cytometric analysis and tumorsphere-forming assays were used to detect the killing effects of LR004-VC-MMAE on cancer stem cells in MDA-MB-468 and MDA-MB-231 cells. The mouse xenograft models were also used to evaluate the antitumor efficacy of LR004-VC-MMAE in vivo. Briefly, BALB/c nude mice were subcutaneously inoculated with MDA-MB-468 or MDA-MB-231 cells. Then they were randomly divided into 4 groups (n = 6 per group) and treated with PBS, naked LR004 (10 mg/kg), LR004-VC-MMAE (10 mg/kg), or doxorubicin, respectively. Tumor sizes and the body weights of mice were measured every 4 days. The effects of LR004-VC-MMAE on apoptosis and cell cycle distribution were analyzed by flow cytometry. Western blotting was used to detect the effects of LR004-VC-MMAE on EGFR, ERK, MEK phosphorylation and tumor stemness marker gene expression. RESULTS: LR004-VC-MMAE with a DAR of 4.02 were obtained. The expression of EGFR was found to be significantly higher in TNBC cells compared with non-TNBC cells (P < 0.01). LR004-VC-MMAE inhibited the proliferation of EGFR-positive TNBC cells, and the IC50 values of MDA-MB-468 and MDA-MB-231 cells treated with LR004-VC-MMAE for 72 h were (0.13 ± 0.02) nmol/L and (0.66 ± 0.06) nmol/L, respectively, which were significantly lower than that of cells treated with MMAE [(3.20 ± 0.60) nmol/L, P < 0.01, and (6.60 ± 0.50) nmol/L, P < 0.001]. LR004-VC-MMAE effectively inhibited migration and invasion of MDA-MB-468 and MDA-MB-231 cells. Moreover, LR004-VC-MMAE also killed tumor stem cells in EGFR-positive TNBC cells and impaired their tumorsphere-forming ability. In TNBC xenograft models, LR004-VC-MMAE at 10 mg/kg significantly suppressed tumor growth and achieved complete tumor regression on day 36. Surprisingly, tumor recurrence was not observed until the end of the experiment on day 52. In a mechanistic study, we found that LR004-VC-MMAE significantly induced cell apoptosis and cell cycle arrest at G2/M phase in MDA-MB-468 [(34 ± 5)% vs. (12 ± 2)%, P < 0.001] and MDA-MB-231 [(27 ± 4)% vs. (18 ± 3)%, P < 0.01] cells. LR004-VC-MMAE also inhibited the activation of EGFR signaling and the expression of cancer stemness marker genes such as Oct4, Sox2, KLF4 and EpCAM. CONCLUSIONS: LR004-VC-MMAE showed effective antitumor activity by inhibiting the activation of EGFR signaling and the expression of cancer stemness marker genes. It might be a promising therapeutic candidate and provides a potential therapeutic avenue for the treatment of EGFR-positive TNBC.


Subject(s)
Immunoconjugates , Triple Negative Breast Neoplasms , Animals , Humans , Mice , ErbB Receptors/metabolism , ErbB Receptors/therapeutic use , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Mice, Nude , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
7.
Cancer Biol Ther ; 21(4): 332-343, 2020 04 02.
Article in English | MEDLINE | ID: mdl-31906826

ABSTRACT

Gemcitabine (Gem) is currently used as the first-line therapy for liver and pancreatic cancer but has limited efficacy in most cases. Dexamethasone (Dex) have been applied as a chemoprotectant and chemosensitizer in cancer chemotherapy. This study further explored the potential of combination of Gem and Dex and tested the hypothesis that glucocorticoid receptor signaling is essential for the synergistic antitumor activity. In the HepG2 and AsPC-1 xenograft models, the combination treatment showed a significantly synergistic antitumor activity. Immunohistochemistry of post-treatment tumors showed a significant decrease in proliferation and angiogenesis as compared to either of the treatments alone. Dex alone and the combination with Gem inhibited the expression of glucocorticoid receptor. The combination of Dex and Gem showed synergistic cytotoxicity in cell lines in vitro. The antiproliferative synergism is prevented by used glucocorticoid receptor (GR) small interfering RNA, demonstrating that the glucocorticoid receptor is required for the antiproliferative synergism of Gem and Dex. The inhibition of glucocorticoid receptor signaling pathway and induction of apoptosis via activation of caspases 3, 8 and 9, PARP, contributed to the synergistic effect of this combination therapy. These results demonstrate that Dex could potentiate the antitumor efficacy of Gem. The synergistic antitumor activity of the combination of Dex and Gem was through glucocorticoid receptor signaling. Taken together, a combination of Dex and Gem shows a significant synergistic antitumor activity and lesser toxicity both in vitro and in vivo and could be a combination chemotherapy for the treatment of highly expression of glucocorticoid receptor patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/pathology , Pancreatic Neoplasms/drug therapy , Receptors, Glucocorticoid/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dexamethasone/administration & dosage , Female , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Gemcitabine
8.
Mol Oncol ; 13(2): 246-263, 2019 02.
Article in English | MEDLINE | ID: mdl-30372581

ABSTRACT

Epidermal growth factor receptor (EGFR) is a rational target for cancer therapy, because its overexpression plays an important oncogenic role in a variety of solid tumors; however, EGFR-targeted antibody-drug conjugate (ADC) therapy for esophageal squamous cell carcinoma (ESCC) is exceedingly rare. LR004 is a novel anti-EGFR antibody with the advantages of improved safety and fewer hypersensitivity reactions. It may be of great value as a carrier in ADCs with high binding affinity and internalization ability. Here, we prepared an EGFR-targeting ADC, LR004-VC-MMAE, and evaluated its antitumor activities against ESCC and EGFR-positive cells. LR004 was covalently conjugated with monomethyl auristatin E (MMAE) via a VC linker by antibody interchain disulfide bond reduction. VC-MMAE was conjugated with LR004 with approximately 4.0 MMAE molecules per ADC. LR004-VC-MMAE showed a potent antitumor effect against ESCC and other EGFR-positive cells with IC50 values of nM concentrations in vitro. The in vivo antitumor effects of LR004-VC-MMAE were investigated in ESCC KYSE520 and A431 xenograft nude mice models. Significant activity was seen at 5 mg·kg-1 , and complete tumor regression was observed at 15 mg·kg-1 in the KYSE520 xenograft nude mice after four injections, while the naked antibody LR004 had little effect on inhibiting tumor growth. Similar promising results were obtained in the A431 models. In addition, the tumors also remained responsive to LR004-VC-MMAE for large tumor experiments (tumor volume 400-500 mm3 ). The study results demonstrated that LR004-VC-MMAE could be a potential therapeutic agent for ESCC and other EGFR-expressing malignancies. We also evaluated PK profile of LR004-VC-MMAE ADC in the mice model, which would provide qualitative guiding significance for the further research.


Subject(s)
Antibodies, Monoclonal/therapeutic use , ErbB Receptors/metabolism , Esophageal Squamous Cell Carcinoma/drug therapy , Immunoconjugates/therapeutic use , Oligopeptides/therapeutic use , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Endocytosis/drug effects , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Inhibitory Concentration 50 , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Oligopeptides/pharmacology , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Acta Pharmacol Sin ; 39(11): 1777-1786, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30013033

ABSTRACT

Defensins play an essential role in innate immunity. In this study, a novel recombinant ß-defensin that targets the epidermal growth factor receptor (EGFR) was designed and prepared. The EGFR-targeting ß-defensin consists of an EGF-derived oligopeptide (Ec), a ß-defensin-1 peptide (hBD1) and a lidamycin-derived apoprotein (LDP), which serves as the "scaffold" for the fusion protein (Ec-LDP-hBD1). Ec-LDP-hBD1 effectively bound to EGFR highly expressed human epidermoid carcinoma A431 cells. The cytotoxicity of Ec-LDP-hBD1 to EGFR highly expressed A431 cells was more potent than that to EGFR low-expressed human lung carcinoma A549 and H460 cells (the IC50 values in A431, A549, and H460 cells were 1.8 ± 0.55, 11.9 ± 0.51, and 5.19 ± 1.21 µmol/L, respectively); in addition, the cytotoxicity of Ec-LDP-hBD1 was much stronger than that of Ec-LDP and hBD1. Moreover, Ec-LDP-hBD1 suppressed cancer cell proliferation and induced mitochondria-mediated apoptosis. Its in vivo anticancer action was evaluated in athymic mice with A431 and H460 xenografts. The mice were administered Ec-LDP-hBD1 (5, 10 mg/kg, i.v.) two times with a weekly interval. Administration of Ec-LDP-hBD1 markedly inhibited the tumor growth without significant body weight changes. The in vivo imaging further revealed that Ec-LDP-hBD1 had a tumor-specific distribution with a clear image of localization. The results demonstrate that the novel recombinant EGFR-targeting ß-defensin Ec-LDP-hBD1 displays both selectivity and enhanced cytotoxicity against relevant cancer cells by inducing mitochondria-mediated apoptosis and exhibits high therapeutic efficacy against the EGFR-expressed carcinoma xenograft. This novel format of ß-defensin, which induces mitochondrial-mediated apoptosis, may play an active role in EGFR-targeting cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Mitochondria/metabolism , Recombinant Fusion Proteins/therapeutic use , beta-Defensins/therapeutic use , Aminoglycosides/metabolism , Aminoglycosides/therapeutic use , Animals , Antineoplastic Agents/metabolism , Apoproteins/metabolism , Apoproteins/therapeutic use , Cell Line, Tumor , Enediynes/metabolism , Enediynes/therapeutic use , ErbB Receptors/metabolism , Female , Humans , Mice, Nude , Mitochondria/pathology , Protein Binding , Recombinant Fusion Proteins/metabolism , Xenograft Model Antitumor Assays , beta-Defensins/metabolism
10.
Oncotarget ; 8(16): 27286-27299, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28460483

ABSTRACT

Epidermal growth factor receptor (EGFR) and insulin-like growth factor 1 receptor (IGF-1R) both overexpressed on non-small cell lung cancer (NSCLC) and are known cooperatively to promote tumor progression and drug resistance. This study was to construct a novel bispecific fusion protein EGF-IGF-LDP-AE consisting of EGFR and IGF-IR specific ligands (EGF and IGF-1) and lidamycin, an enediyne antibiotic with potent antitumor activity, and investigate its antitumor efficacy against NSCLC. Binding and internalization assays showed that EGF-IGF-LDP protein could bind to NSCLC cells with high affinity and then internalized into cells with higher efficiency than that of monospecific proteins. In vitro, the enediyne-energized analogue of bispecific fusion protein (EGF-IGF-LDP-AE) displayed extremely potent cytotoxicity to NSCLC cell lines with IC50<10-11 mol/L. Moreover, the bispecific protein EGF-IGF-LDP-AE was more cytotoxic than monospecific proteins (EGF-LDP-AE and LDP-IGF-AE) and lidamycin. In vivo, EGF-IGF-LDP-AE markedly inhibited the growth of A549 xenografts, and the efficacy was more potent than that of lidamycin and monospecific counterparts. EGF-IGF-LDP-AE caused significant cell cycle arrest and it also induced cell apoptosis in a dosage-dependent manner. Pretreatment with EGF-IGF-LDP-AE inhibited EGF-, IGF-stimulated EGFR and IGF-1R phosphorylation, and blocked two main downstream signaling molecules AKT and ERK activation. These data suggested that EGF-LDP-IGF-AE protein would be a promising targeted agent for NSCLC patients with EGFR and/or IGF-1R overexpression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Enediynes , ErbB Receptors/antagonists & inhibitors , Insulin-Like Growth Factor I/antagonists & inhibitors , Lung Neoplasms/metabolism , Recombinant Fusion Proteins/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Enediynes/chemistry , ErbB Receptors/metabolism , Female , Humans , Insulin-Like Growth Factor I/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(36): 58418-58434, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27517152

ABSTRACT

K-Ras mutant pancreatic cancer cells display intensive macropinocytosis, indicating that this process may be exploited in the design of anticancer targeted therapies. In this study, we constructed a macropinocytosis-oriented recombinantly tailored defensin (DF-HSA) which consists of human ß-defensin-2 (DF) and human serum albumin (HSA). The macropinocytosis intensity and cytotoxicity of DF-HSA were investigated in K-Ras mutant MIA PaCa-2 cells and wild-type BxPC-3 cells. As found, the DF-HSA uptake in MIA PaCa-2 cells was much higher than that in wild-type BxPC-3 cells. Correspondingly, the cytotoxicity of DF-HSA to MIA PaCa-2 cells was more potent than that to BxPC-3 cells. In addition, the cytotoxicity of DF-HSA was much stronger than that of ß-defensin HBD2. DF-HSA suppressed cancer cell proliferation and induced mitochondrial pathway apoptosis. Notably, DF-HSA significantly inhibited the growth of human pancreatic carcinoma MIA PaCa-2 xenograft in athymic mice at well tolerated dose. By in vivo imaging, DF-HSA displayed a prominent accumulation in the tumor. The study indicates that the recombinantly tailored ß-defensin can intensively enter into the K-Ras mutant pancreatic cancer cells through macropinocytosis-mediated process and exert potent therapeutic efficacy against the pancreatic carcinoma xenograft. The novel format of ß-defensin may play an active role in macropinocytosis-mediated targeting therapy.


Subject(s)
Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pinocytosis , Proto-Oncogene Proteins p21(ras)/genetics , Serum Albumin, Human/metabolism , beta-Defensins/metabolism , A549 Cells , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , Genes, ras , Genetic Vectors , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasm Transplantation , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism
12.
Eur J Pharm Sci ; 47(4): 781-9, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-22982402

ABSTRACT

Arginine-rich peptides belong to a subclass of cell penetrating peptides that are taken up by living cells and can be detected freely diffusing inside the cytoplasm and nucleoplasm. This phenomenon has been attributed to either an endocytotic mode of uptake and a subsequent release from vesicles or a direct membrane penetration. Lidamycin is an antitumor antibiotic, which consists of an active enediyne chromophore (AE) and a noncovalently bound apoprotein (LDP). In the present study, a fusion protein (Arg)(9)-LDP composed of cell penetrating peptide (Arg)(9) and LDP was prepared by DNA recombination, and the enediyne-energized fusion protein (Arg)(9)-LDP-AE was prepared by molecular reconstitution. The data in fixed cells demonstrated that (Arg)(9)-LDP could rapidly enter cells, and the results based on fluorescence activated cell sorting indicated that the major route for (Arg)(9)-mediated cellular uptake of protein molecules was endocytosis. (Arg)(9)-LDP-AE demonstrated more potent cytotoxicity against different carcinoma cell lines than lidamycin in vitro. In the mouse hepatoma 22 model, (Arg)(9)-LDP-AE (0.3mg/kg) suppressed the tumor growth by 89.2%, whereas lidamycin (0.05 mg/kg) by 74.6%. Furthermore, in the glioma U87 xenograft model in nude mice, (Arg)(9)-LDP-AE at 0.2mg/kg suppressed tumor growth by 88.8%, compared with that of lidamycin by 62.9% at 0.05 mg/kg. No obvious toxic effects were observed in all groups during treatments. The results showed that energized fusion protein (Arg)(9)-LDP-AE was more effective than lidamycin and would be a promising candidate for glioma therapy. In addition, this approach to manufacturing fusion proteins might serve as a technology platform for the development of new cell penetrating peptides-based drugs.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoproteins/pharmacology , Cell-Penetrating Peptides/pharmacology , Enediynes/pharmacology , Recombinant Fusion Proteins/pharmacology , Aminoglycosides/pharmacology , Animals , Antibiotics, Antineoplastic/metabolism , Apoproteins/genetics , Apoproteins/metabolism , Arginine/genetics , Arginine/metabolism , Base Sequence , Cell Line, Tumor , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/metabolism , Endocytosis/drug effects , Endocytosis/physiology , Enediynes/metabolism , Female , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Oncol Rep ; 28(4): 1193-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22797730

ABSTRACT

Gelatinases play an important role in tumor growth and metastasis, and overexpression of these molecules is strongly correlated with poor prognosis in a variety of malignant tumors. Lidamycin is an enediyne antitumor antibiotic with potent cytotoxicity. We previously reported that a tandem scFv format (dFv-LDP-AE) showed enhanced binding ability with gelatinases compared with the scFv-lidamycin conjugate (Fv-LDP-AE). In this study, the antitumor activities of dFv-LDP-AE on hepatocellular carcinoma (HCC) were evaluated in vitro and in vivo. By SDS-PAGE analysis, it was found that partial fusion protein dFv-LDP existed as dimer; the results of ELISA and immunofluorescence demonstrated that the fusion protein dFv-LDP could efficiently bind to hepatoma cells in vitro. The apparent arrest of cell cycle at G2/M phase and induction of apoptosis at nanomole levels indicated that the dFv-LDP-AE was very potent against HCC. In in vivo experiments, dFv-LDP-AE shown enhanced cytotoxic effects compared to those of LDM. Administration at mouse tolerable dosage level, the inhibition rate of tumor growth was 89.5% of dFv-LDP-AE vs. 73.6% of LDM on transplantable H22 in mice (P<0.05) and, 87.3% of dFv-LDP-AE vs. 63.4% of LDM on hepatoma Bel-7402 in athymic mice (P<0.01). Small animal optical imaging showed that the FITC-labeled dFv-LDP preferentially localized in the tumor site in less than 30 min, which demonstrated remarkable tumor-targeting properties. Taken together with the above findings, the enediyne-energized fusion protein dFv-LDP-AE showed potential application as a new agent for therapeutic appications in HCC.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Enediynes/pharmacology , Gelatinases/immunology , Single-Chain Antibodies/pharmacology , Aminoglycosides/metabolism , Animals , Antibiotics, Antineoplastic/immunology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Enediynes/metabolism , Gelatinases/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Male , Mice , Mice, Nude , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/immunology , Xenograft Model Antitumor Assays
14.
Yao Xue Xue Bao ; 47(10): 1261-8, 2012 Oct.
Article in Chinese | MEDLINE | ID: mdl-23289136

ABSTRACT

The use of monoclonal antibodies (mAbs) for cancer therapy has achieved considerable success in recent years. Approximate 17 monoclonal antibodies have been approved as cancer therapeutics since 1997. Antibody-drug conjugates (ADC) are powerful new treatment options for cancer, and naked antibodies have recently achieved remarkable success. The safety and effectiveness of therapeutic mAbs in oncology vary depending on the nature of the target antigen and the mechanisms of tumor cell killing. This review provides a summary of the current state of antibody-based cancer therapy, including the mechanisms of tumor cell killing by antibodies, tumor antigens as antibody targets, clinical effectiveness of antibodies in cancer patients and nanoparticles-based ADCs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/therapy , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Humans , Nanoparticles , Neoplasms/immunology
15.
Yao Xue Xue Bao ; 44(6): 597-602, 2009 Jun.
Article in Chinese | MEDLINE | ID: mdl-19806889

ABSTRACT

Recent studies have shown that epidermal growth factor receptor (EGFR) is an important target for cancer therapy. The present study prepared single chain Fv (scFv) directed against EGFR. Balb/c mice were immunized by human carcinoma A431 cells, and total RNA of the splenic cells was extracted. VH and VL gene fragments were amplified by RT-PCR and further joined into scFv gene with a linker, then scFv gene fragments were ligated into the phagemid vector pCANTAB 5E. The phagemid containing scFv were transformed into electro-competent E. coli TG1 cells. The recombinant phage antibody library was constructed through rescuing the transformed cells with help phage M13K07. The specified recombinant phages were enriched through 5 rounds of affinity panning and the anti-EGFR phage scFv clones were screened and identified with ELISA. A total of 48 clones from the library were selected randomly and 45 clones were identified positive. After infecting E. coli HB2151 cells with one positive clone, soluble recombinant antibodies about 27 kD were produced and located in the periplasm and the supernatant. The result of sequencing showed that the scFv gene was 768 bp, which encoded 256 amino acid residues. VH and VL including 3 CDRs and 4 FRs, respectively, were all homologous to mouse Ig. The soluble scFv showed the specific binding activity to purified EGFR and EGFR located in carcinoma cell membrane. The successful preparation of anti-EGFR scFv will provide an EGFR targeted molecule for the development of antibody-based drugs and biological therapy of cancer.


Subject(s)
ErbB Receptors/genetics , Immunoglobulin Light Chains/genetics , Single-Chain Antibodies/genetics , Animals , Antibodies, Monoclonal , Cell Line, Tumor , ErbB Receptors/immunology , Humans , Immunoglobulin Light Chains/immunology , Male , Mice , Mice, Inbred BALB C , Peptide Library , Single-Chain Antibodies/immunology
16.
Zhonghua Yi Xue Za Zhi ; 89(20): 1411-5, 2009 May 26.
Article in Chinese | MEDLINE | ID: mdl-19671337

ABSTRACT

OBJECTIVE: To study the effect of anti-type IV collagenase single-chain antibody scFv (3G11) on bleomycin (BLM)-induced pulmonary fibrosis. METHODS: C57BL/6 mice were divided into six groups (blank, saline control, BLM, low-dose treatment, intermediate-dose treatment, and high-dose treatment), the animal model of pulmonary fibrosis was induced with 400 ng/g intratracheal BLM. The day that treatment groups were injected with BLM was marked Day 0. Then at Days 1 - 7, scFv (3G11) was injected once intraperitoneally each day with three dosage [low-dose treatment (15 microg/g), intermediate-dose treatment (30 microg/g) and high-dose treatment (45 microg/g)]. At Day 21, 6 mice of each group (saline control, BLM, intermediate-dose treatment) were sacrificed and pathomorphological changes of left lungs evaluated with HE stained sections. Meanwhile, 6 mice of each group (blank, saline control, BLM, low-dose treatment, intermediate-dose treatment, and high-dose treatment) were sacrificed, the content of hydroxyproline was detected to measure the degree of collagen deposition. Macrophages were extracted from murine abdominal cavity and primarily cultured. And the inhibition of matrix metalloproteinase (MMP)-2 and MMP-9 excretion by different concentrations of scFv (3G11) (0, 15, 30, 45, and 60 micromol/L) was determined by gelatin zymography. RESULTS: By image analysis, the degree of fibrosis in the lungs of treated group was lighter than that of the BLM group. The percentages of the area of interalveolar septum (45.3% +/- 3.2%) and the number of nucleated cells (451 +/- 47) of treated group were remarkably reduced as compared with the BLM group (59.0% +/- 3.0%, 599 +/- 42, both P < 0.01). The content of hydroxyproline of the intermediate-dose treatment group [(0.82 +/- 0.05) microg/mg] and high-dose treatment group [(0.80 +/- 0.03) microg/mg] was lower than that of the BLM group [(0.92 +/- 0.07) microg/mg, P < 0.05, P < 0.01]. The result of gelatin zymography demonstrated that the excretion of MMP-2 and MMP-9 by macrophage was inhibited by scFv (3G11) at the concentration of 15 micromol/L. CONCLUSION: Single-chain antibody directed against type IV collagenase might inhibit the development of bleomycin-induced pulmonary fibrosis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lung/pathology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Animals , Bleomycin/adverse effects , Immunoglobulin Variable Region/therapeutic use , Lung/metabolism , Male , Matrix Metalloproteinase 2/immunology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced
17.
Ai Zheng ; 28(6): 561-8, 2009 Jun.
Article in Chinese | MEDLINE | ID: mdl-19635191

ABSTRACT

BACKGROUND AND OBJECTIVE: Epidermal growth factor receptor (EGFR) is abnormally overexpressed on many kinds of tumor cells. Lidamycin is an enediyne antibiotic with highly potent antitumor activity. This study was to construct a novel fusion protein by recombining EGFR specific oligopeptide ligand and lidamycin, and investigate its antitumor efficacy. METHODS: The fusion protein (Ec-LDP) was expressed in E.coli and purified by affinity chromatography. The purity of Ec-LDP was analyzed by high performance liquid chromatography (HPLC). ELISA, flow cytometry (FCM) and immunofluorescence assay were used to analyze the binding activity of Ec-LDP to different cancer cell lines. The energized fusion protein Ec-LDP-AE was prepared by integrating the active enediyne chromophore (AE) of lidamycin into the Ec-LDP protein. The cytotoxicity of Ec-LDP-AE was measured by MTT assay. RESULTS: Ec-LDP fusion protein was successfully constructed and secretorily expressed in E.coli, and the production of Ec-LDP protein was 18 mg per liter fermentation broth. The purity of Ec-LDP protein was 95.3%. Ec-LDP protein had strong binding activity to cancer cell lines highly expressing EGFR, such as MCF-7 and A431 cells. However, Ec-LDP had no binding activity to EGFR negative NIH 3T3 cells. The energized fusion protein Ec-LDP-AE showed potent cytotoxicity to MCF-7 and A431 cells with the half maximal inhibitory concentration (IC50) values of 3.06 x 10(-11) mol/L and 9.38 x 10(-13) mol/L, respectively. CONCLUSION: The energized fusion protein Ec-LDP-AE binds to EGFR specifically and kills cancer cells efficiently.


Subject(s)
Aminoglycosides/pharmacology , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Enediynes/pharmacology , ErbB Receptors/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , ErbB Receptors/genetics , Female , Humans , Ligands , Mice , NIH 3T3 Cells , Oligopeptides/genetics , Oligopeptides/pharmacology , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology
18.
Anticancer Drugs ; 18(2): 127-37, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17159599

ABSTRACT

Single-domain antibodies are attractive as tumor-targeting vehicles because of their much smaller size than intact antibody molecules. Lidamycin is a macromolecular antitumor antibiotic, which consists of a labile enediyne chromophore (AE) and a noncovalently bound apoprotein (LDP). An enediyne-energized fusion protein VH-LDP-AE composed of single-domain antibody directed against type IV collagenase and lidamycin was prepared by a novel two-step method including DNA recombination and molecular reconstitution. VH-LDP-AE demonstrated extremely potent cytotoxicity to cancer cells and marked antiangiogenic activity in vitro. In the mouse hepatoma 22 model, drugs were administered intravenously as a single dose on day 1 with maximal tolerated doses. VH-LDP-AE (0.25 mg/kg) suppressed the tumor growth by 95.9%, whereas lidamycin (0.05 mg/kg) and mitomycin (1 mg/kg) by 79.6 and 51.1%, respectively. In the HT-1080 xenograft model in nude mice, drugs were given intravenously as a single dose on day 4 after tumor implantation. VH-LDP-AE at 0.25 mg/kg suppressed tumor growth by 76% (P<0.05) compared with that of lidamycin at 0.05 mg/kg (53%) on day 18. No obvious toxic effects were observed in all groups during treatments. The results showed that energized fusion protein VH-LDP-AE was more effective than lidamycin and mitomycin. These properties, together with its much smaller size than conventional antibody-based agents, suggested that VH-LDP-AE would be a promising candidate for cancer-targeting therapy. In addition, the two-step approach could serve as a new technology platform for making a series of highly potent engineered antibody-based drugs for a variety of cancers.


Subject(s)
Antibodies/chemistry , Antibodies/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Allantois/chemistry , Allantois/drug effects , Amino Acid Sequence , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Blotting, Western , Cell Line, Tumor , Chick Embryo , Collagenases/chemistry , Collagenases/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors , Indicators and Reagents , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Microtubules , Molecular Sequence Data , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...