Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Pharmaceuticals (Basel) ; 16(12)2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38139816

ABSTRACT

This Special Issue focuses on the use of therapeutic antibodies in vitro, in vivo, and in clinical studies [...].

2.
Sci Rep ; 13(1): 16017, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37749105

ABSTRACT

The PI3K-Akt-mTOR (PAM) pathway is implicated in tumor progression in many tumor types, including metastatic gastric cancer (GC). The initial promise of PAM inhibitors has been unrealized in the clinic, presumably due, in part, to the up-regulation of Akt signaling that occurs when the pathway is inhibited. Here we present that DIACC3010 (formerly M2698), an inhibitor of two nodes in the PAM pathway, p70S6K and Akt 1/3, blocks the pathway in in vitro and in vivo preclinical models of GC while providing a mechanism that inhibits signaling from subsequent Akt up-regulation. Utilizing GC cell lines and xenograft models, we identified potential markers of DIACC3010-sensitivity in Her2-negative tumors, i.e., PIK3CA mutations, low basal pERK, and a group of differentially expressed genes (DEGs). The combination of DIACC3010 and trastuzumab was evaluated in Her2-positive cell lines and models. Potential biomarkers for the synergistic efficacy of the combination of DIACC3010 + trastuzumab also included DEGs as well as a lack of up-regulation of pERK. Of 27 GC patient-derived xenograft (PDX) models tested in BALB/c nu/nu mice, 59% were sensitive to DIACC3010 + trastuzumab. Of the 21 HER2-negative PDX models, DIACC3010 significantly inhibited the growth of 38%. Altogether, these results provide a path forward to validate the potential biomarkers of DIACC3010 sensitivity in GC and support clinical evaluation of DIACC3010 monotherapy and combination with trastuzumab in patients with HER2- negative and positive advanced GCs, respectively.


Subject(s)
Stomach Neoplasms , Animals , Mice , Humans , Stomach Neoplasms/drug therapy , Ribosomal Protein S6 Kinases, 70-kDa , Proto-Oncogene Proteins c-akt , Phosphatidylinositol 3-Kinases , Protein Kinase Inhibitors , Angiogenesis Inhibitors , Disease Models, Animal
3.
Mol Pharm ; 20(2): 1156-1167, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36573995

ABSTRACT

Astatine-211 (211At) is an alpha emitter applicable to radioimmunotherapy (RIT), a cancer treatment that utilizes radioactive antibodies to target tumors. In the preparation of 211At-labeled monoclonal antibodies (211At-mAbs), the possibility of radionuclide-induced antibody denaturation (radiolysis) is of concern. Our previous study showed that this 211At-induced radiochemical reaction disrupts the cellular binding activity of an astatinated mAb, resulting in attenuation of in vivo antitumor effects, whereas sodium ascorbate (SA), a free radical scavenger, prevents antibody denaturation, contributing to the maintenance of binding and antitumor activity. However, the influence of antibody denaturation on the pharmacokinetics of 211At-mAbs relating to tumor accumulation, blood circulation time, and distribution to normal organs remains unclear. In this study, we use a radioactive anti-human epidermal growth factor receptor 2 (anti-HER2) mAb to demonstrate that an 211At-induced radiochemical reaction disrupts active targeting via an antigen-antibody interaction, whereas SA helps to maintain targeting. In contrast, there was no difference in blood circulation time as well as distribution to normal organs between the stabilized and denatured immunoconjugates, indicating that antibody denaturation may not affect tumor accumulation via passive targeting based on the enhanced permeability and retention effect. In a high-HER2-expressing xenograft model treated with 1 MBq of 211At-anti-HER2 mAbs, SA-dependent maintenance of active targeting contributed to a significantly better response. In treatment with 0.5 or 0.2 MBq, the stabilized radioactive mAb significantly reduced tumor growth compared to the denatured immunoconjugate. Additionally, through a comparison between a stabilized 211At-anti-HER2 mAb and radioactive nontargeted control mAb, we demonstrate that active targeting significantly enhances tumor accumulation of radioactivity and in vivo antitumor effect. In RIT with 211At, active targeting contributes to efficient tumor accumulation of radioactivity, resulting in a potent antitumor effect. SA-dependent protection that successfully maintains tumor targeting will facilitate the clinical application of alpha-RIT.


Subject(s)
Immunoconjugates , Neoplasms , Humans , Antibodies, Monoclonal , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radioisotopes , Radioimmunotherapy/methods , Cell Line, Tumor
4.
Pharmaceuticals (Basel) ; 15(2)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35215338

ABSTRACT

Photoimmunotherapy (PIT) is a promising tumor-selective treatment method that uses light-absorbing dye-conjugated antibodies and light irradiation. It has been reported that IR700 fluorescence changes with light irradiation. The purpose of this study was to investigate the fluorescence intensity and antitumor effect of PIT using real-time fluorescence observation of tumors and predict the required irradiation dose. The near-infrared camera system LIGHTVISION was used to image IR700 during PIT treatment. IR700 showed a sharp decrease in fluorescence intensity in the early stage of treatment and almost reached a plateau at an irradiation dose of 40 J/cm. Cetuximab-PIT for A431 xenografts was performed at multiple doses from 0-100 J/cm. A significant antitumor effect was observed at 40 J/cm compared to no irradiation, and there was no significant difference between 40 J/cm and 100 J/cm. These results suggest that the rate of decay of the tumor fluorescence intensity correlates with the antitumor effect by real-time fluorescence imaging during PIT. In addition, when the fluorescence intensity of the tumor plateaued in real-time fluorescence imaging, it was assumed that the laser dose was necessary for treatment.

5.
ACS Omega ; 6(23): 14887-14895, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34151070

ABSTRACT

211At, an α-particle emitter, has recently attracted attention for radioimmunotherapy of intractable cancers. However, our sodium dodecyl sulfate polyacrylamide gel electrophoresis and flow cytometry analyses revealed that 211At-labeled immunoconjugates are easily disrupted. Luminol assay revealed that reactive oxygen species generated from radiolysis of water caused the disruption of 211At-labeled immunoconjugates. To retain their functions, we explored methods to protect 211At-immunoconjugates from oxidation and enhance their stability. Among several other reducing agents, sodium ascorbate most safely and successfully protected 211At-labeled trastuzumab from oxidative stress and retained the stability of the 211At-labeled antibody and its cytotoxicity against antigen-expressing cells for several days.

6.
Clin Transl Gastroenterol ; 12(3): e00319, 2021 03.
Article in English | MEDLINE | ID: mdl-33939384

ABSTRACT

INTRODUCTION: Noninvasive colorectal cancer (CRC) screening methods with higher sensitivity for advanced colorectal neoplasia (ACN) than the fecal immunochemical test (FIT) alone are warranted. This study aimed to elucidate the diagnostic performance of a risk stratification score calculated using baseline individual characteristics and its combination with FIT for detecting ACN. METHODS: This cross-sectional analysis of data from a prospective cohort in Izu Oshima, Japan, included asymptomatic individuals age 40-79 years who underwent both 2-day quantitative FIT and screening colonoscopy. The 8-point risk score, calculated based on age, sex, CRC family history, body mass index, and smoking history, was assessed. Colonoscopy results were used as reference. RESULTS: Overall, 1,191 individuals were included, and 112 had ACN. The sensitivity and specificity of the 1-/2-day FIT (cutoff: 50-200 ng Hb/mL) for ACN were 17.9%-33.9% (4.9%-22.0% for right-sided ACN) and 91.8%-97.6%, respectively. The risk score's c-statistic for ACN was 0.66, and combining the score (cutoff: 5 points) with 1-/2-day FIT (cutoff: 50-200 ng Hb/mL) yielded a sensitivity and specificity for ACN of 46.4%-56.3% (43.9%-48.8% for right-sided ACN) and 76.6%-80.8%, respectively. The specificity of the risk score and FIT combination for all adenomatous lesions was 82.4%-86.4%. DISCUSSION: The 8-point risk score remarkably increased the sensitivity for ACN, particularly for right-sided ACN. Although the specificity decreased, it was still maintained at a relatively high level. The risk score and FIT combination has the potential to become a viable noninvasive CRC screening option.


Subject(s)
Colorectal Neoplasms/diagnosis , Early Detection of Cancer/methods , Mass Screening/methods , Adult , Aged , Asymptomatic Diseases/epidemiology , Colonoscopy/statistics & numerical data , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/prevention & control , Cross-Sectional Studies , Female , Humans , Japan/epidemiology , Male , Middle Aged , Prospective Studies , Risk Assessment/methods , Risk Factors , Sensitivity and Specificity
7.
BMC Cancer ; 21(1): 302, 2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33757462

ABSTRACT

BACKGROUND: Transmembrane protein 180 (TMEM180) is a newly identified colorectal cancer (CRC)-specific molecule that is expressed very rarely in normal tissue and up-regulated under hypoxic conditions. We developed a monoclonal antibody (mAb) against TMEM180 and decided to examine the medical significance using the mAb. METHODS: A total of 157 patients (86 men and 71 women; median age 63.0 years) with stage III CRC who underwent curative surgery were analyzed for TMEM180 expression as a retrospective cohort design. Immunohistochemistry with anti-TMEM180 mAb was conducted on frozen sections, and the data were evaluated for any correlation with clinicopathological indices or prognosis. SW480 CRC cells were examined to investigate the relationship between the expression of TMEM180 and tumourigenesis of xenografts. RESULTS: In total, 92 cases had low TMEM expression and 65 had high TMEM180 expression. For disease-free survival, hazard ratio in high-TMEM180 cases was 1.449 (95% confidential interval = 0.802-2.619) higher than in low-TMEM180 cases, but the difference was not significant (p = 0.219). For cancer specific survival, hazard ratio in high-TMEM180 cases was 3.302 (95% confidential interval = 1.088-10.020), significantly higher than in low-TMEM180 cases (p = 0.035). In an assay examining in vitro colony-forming activity in soft agar, SW480-WT cells clearly formed colonies, but neither KD1 nor KD2 cells did. The in vivo tumour-initiating activity of SW480 cell lines was positively correlated with the level of TMEM180 expression. CONCLUSION: These results indicate that TMEM180 is a useful marker for clinical prognosis in patients with CRC. We believe that these fundamental data warrant further basic and translational studies of TMEM180, and its mAb, for development of therapeutics against CRC.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/mortality , Membrane Proteins/analysis , Aged , Cell Line, Tumor , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Neoplasm Staging , Retrospective Studies
8.
Cancer Sci ; 112(5): 1975-1986, 2021 May.
Article in English | MEDLINE | ID: mdl-33606344

ABSTRACT

Tissue factor (TF), the trigger protein of the extrinsic blood coagulation cascade, is abundantly expressed in various cancers including gastric cancer. Anti-TF monoclonal antibodies (mAbs) capable of targeting cancers have been successfully applied to armed antibodies such as antibody-drug conjugates (ADCs) and molecular imaging probes. We prepared an anti-TF mAb, clone 1084, labeled with astatine-211 (211 At), as a promising alpha emitter for cancer treatment. Alpha particles are characterized by high linear energy transfer and a range of 50-100 µm in tissue. Therefore, selective and efficient tumor accumulation of alpha emitters results in potent antitumor activities against cancer cells with minor effects on normal cells adjacent to the tumor. Although the 211 At-conjugated clone 1084 (211 At-anti-TF mAb) was disrupted by an 211 At-induced radiochemical reaction, we demonstrated that astatinated anti-TF mAbs eluted in 0.6% or 1.2% sodium ascorbate (SA) solution were protected from antibody denaturation, which contributed to the maintenance of cellular binding activities and cytocidal effects of this immunoconjugate. Although body weight loss was observed in mice administered a 1.2% SA solution, the loss was transient and the radioprotectant seemed to be tolerable in vivo. In a high TF-expressing gastric cancer xenograft model, 211 At-anti-TF mAb in 1.2% SA exerted a significantly greater antitumor effect than nonprotected 211 At-anti-TF mAb. Moreover, the antitumor activities of the protected immunoconjugate in gastric cancer xenograft models were dependent on the level of TF in cancer cells. These findings suggest the clinical availability of the radioprotectant and applicability of clone 1084 to 211 At-radioimmunotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Ascorbic Acid/therapeutic use , Astatine/therapeutic use , Immunoconjugates/therapeutic use , Radioimmunotherapy/methods , Stomach Neoplasms/therapy , Thromboplastin/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Astatine/pharmacokinetics , Blood Coagulation/physiology , Body Weight , Cell Line, Tumor , Female , Heterografts , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Linear Energy Transfer , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Denaturation , Radiation-Protective Agents/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Thromboplastin/metabolism
9.
Oncol Rep ; 45(1): 329-336, 2021 01.
Article in English | MEDLINE | ID: mdl-33200231

ABSTRACT

Tissue factor (TF) is an attractive target for cancer therapy due to its overexpression in multiple types of malignancies. In addition, TF has been reported to play functional roles in both cancer development and metastasis. Several groups have already developed antibody­drug conjugates (ADCs) against TF for use as cancer treatments, and have demonstrated their efficacies in conventional subcutaneous xenograft models and patient­derived xenograft models. However, no previous studies have investigated the effectiveness of anti­TF ADC in an advanced­stage cancer model. The present study developed an original humanized anti­TF monoclonal antibody conjugated with monomethyl auristatin E, and evaluated its in vivo efficacy in a pancreatic cancer xenograft model with peritoneal dissemination. In vitro assays demonstrated that the anti­TF ADC had potent binding affinity and cytotoxic activity against human pancreatic cancer cells that strongly expressed TF antigens. The anti­TF ADC also exhibited greater antitumor effect than that of a control ADC in conventional subcutaneous xenograft models, with efficacy depending on the TF expression in the tumor tissues. Furthermore, the anti­TF ADC significantly inhibited tumor growth in an orthotopic xenograft model, and extended the survival period in a murine peritoneal dissemination model. These results indicated that anti­TF ADC has the potential to be an effective treatment not only for primary tumors, but also for those that are widely disseminated. Therefore, it can be concluded that ADC targeting TF may be a promising agent for advanced pancreatic cancer therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunoconjugates/therapeutic use , Pancreatic Neoplasms/drug therapy , Thromboplastin/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Peritoneum/pathology , Xenograft Model Antitumor Assays
10.
Cancer Sci ; 111(9): 3386-3394, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32639672

ABSTRACT

Cell line-derived xenograft (CDX) models created by implanting cancer cell lines into immunodeficient mice have contributed largely to the development of cancer drug therapies. However, cell lines often lose their original biological characteristics through many passages and cancer tissues in CDX models have many cancer cells and few cancer stromal cells, therefore CDX models are currently considered not suitable for predicting the results of clinical studies. Conversely, patient-derived xenograft (PDX) models are gaining importance, as human cancer biological characteristics and microenvironments are recreated by implanting tumor tissue into immunodeficient mice. These highly expected, evidently beneficial PDX models have been used in some basic research and are becoming more generalized. However, quality control and quality assurance criteria have not been established for them, and challenges and problems in the utilization of valuable PDX models in drug development have yet to be clarified. In this report, we conducted a questionnaire survey among researchers in Japanese academic institutions and pharmaceutical companies to understand the current status of PDX models in Japan. Based on the questionnaire results, we summarized the situations surrounding respondent's utilization and quality control in the development of anticancer drugs and proposed several measures to facilitate the utilization of PDX models in the development of anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Xenograft Model Antitumor Assays , Animals , Disease Models, Animal , Drug Development , Drug Screening Assays, Antitumor/methods , Humans , Japan , Mice , Species Specificity
11.
J Control Release ; 323: 138-150, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32259544

ABSTRACT

It has been preclinically and clinically proven that anticancer agent-incorporating (ACA-incorporating) polymeric micelles selectively accumulate in tumor via the enhanced permeability and retention (EPR) effect, yielding a wider therapeutic window and greater safety than conventional low-molecular weight ACAs. To increase the antitumor effect of these safer micelle formulations, epirubicin-incorporating polymer micelles (NC-6300) conjugated with monoclonal antibodies (mAbs) have been prepared. In this study, we used two types of mAb: an anti-tissue factor (TF) mAb that does not exert a direct cytocidal effect, and an anti-HER2 mAb that has a direct cytocidal effect. We compared the antitumor effects and pharmacological properties of the two types of antibody conjugated to NC-6300. Immunomicelles conjugated to anti-TF mAb exerted greater antitumor activity toward TF-positive stomach cancer than the combination of anti-TF mAb and NC-6300, and were distributed more uniformly throughout TF-positive tumor tissue than NC-6300. On the other hand, immunomicelles conjugated to anti-HER2 mAb did not exert significant antitumor activity toward HER2-positive stomach cancer relative to the combined use of anti-HER2 mAb and NC-6300. Thus, this immunomicelle-based strategy may be useful for antibodies that target cancer as pilot molecules even when the antibodies themselves do not have an antitumor effect.


Subject(s)
Antineoplastic Agents , Micelles , Antibiotics, Antineoplastic , Antibodies, Monoclonal , Cell Line, Tumor
12.
Mol Cancer Ther ; 18(11): 2043-2050, 2019 11.
Article in English | MEDLINE | ID: mdl-31395690

ABSTRACT

HER3 is overexpressed in several cancers, including colorectal cancer. Although therapies with anti-HER3 antibodies have been investigated, significant clinical benefits have not been reported. U3-1402 is a novel HER3-antibody-drug conjugate (ADC) composed of the HER3 antibody patritumab and a novel topoisomerase I inhibitor, DX-8951 derivative (DXd). The sensitivity of DXd was evaluated by a growth inhibition assay. The antitumor activity of U3-1402 was evaluated in a murine xenograft model in which its effects on cells, with a range of HER3 expression levels, were compared with those of patritumab alone, irinotecan, control-ADC, and saline. In the growth inhibition assay, all colorectal cancer cell lines were sensitive to DXd. In the tumor xenograft model, significant tumor regression with U3-1402 was observed both in the DiFi cell line (high HER3 expression; KRAS wild type) and in SW620 (high HER3 expression; KRAS mutation), but no treatment effect was observed in Colo320DM (low HER3 expression). Notably, SW620 tumor growth was significantly suppressed with U3-1402 compared with the saline-treated group (P < 0.001) and showed greater activity compared with the irinotecan group. By contrast, patritumab alone, control-ADC, and saline did not significantly differ in tumor growth inhibition. The antitumor activity of U3-1402 was dependent on HER3 expression level, but not on KRAS mutation status. These results support further investigation of development strategies for U3-1402 in patients with HER3-expressing colorectal cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Broadly Neutralizing Antibodies/chemistry , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Immunoconjugates/administration & dosage , Receptor, ErbB-3/metabolism , Animals , Camptothecin/chemistry , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Mice , Receptor, ErbB-3/antagonists & inhibitors , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
13.
Cancer Sci ; 110(10): 3296-3305, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31348600

ABSTRACT

Tissue factor (TF) is known to be overexpressed in various cancers including pancreatic cancer. The upregulation of TF expression has been observed not only in tumor cells, but also in tumor stromal cells. Because of the potential of TF as a delivery target, several studies investigated the effectiveness of Ab-drug conjugates (ADCs) against TF for cancer therapy. However, it is still unclear whether anti-TF ADC can exert toxicity against both tumor cells and tumor stromal cells. Here, we prepared ADC using a rat anti-mouse TF mAb (clone.1157) and 2 types of in vivo murine pancreatic cancer models, one s.c. and other orthotopic with an abundant tumor stroma. We also compared the feasibility of bis-alkylating conjugation (bisAlk) with that of conventional maleimide-based conjugation (MC). In the s.c. models, anti-TF ADC showed greater antitumor effects than control ADC. The results also indicated that the bisAlk linker might be more suitable than the MC linker for cancer treatments. In the orthotopic model, anti-TF ADC showed greater in vivo efficacy and more extended survival time control ADC. Treatment with anti-TF ADC (20 mg/kg, three times a week) did not affect mouse body weight changes in any in vivo experiment. Furthermore, immunofluorescence staining indicated that anti-TF ADC delivered agents not only to TF-positive tumor cells, but also to TF-positive tumor vascular endothelial cells and other tumor stromal cells. We conclude that anti-TF ADC should be a selective and potent drug for pancreatic cancer therapy.


Subject(s)
Alkylating Agents/chemistry , Antineoplastic Agents, Immunological/administration & dosage , Immunoconjugates/administration & dosage , Maleimides/chemistry , Pancreatic Neoplasms/drug therapy , Thromboplastin/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Administration Schedule , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Mice , Mice, Transgenic , Pancreatic Neoplasms/metabolism , Rats , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Xenograft Model Antitumor Assays
14.
Cells ; 8(5)2019 05 06.
Article in English | MEDLINE | ID: mdl-31064068

ABSTRACT

Patient-derived xenograft (PDX) models are used as powerful tools for understanding cancer biology in PDX clinical trials and co-clinical trials. In this systematic review, we focus on PDX clinical trials or co-clinical trials for drug development in solid tumors and summarize the utility of PDX models in the development of anti-cancer drugs, as well as the challenges involved in this approach, following the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Recently, the assessment of drug efficacy by PDX clinical and co-clinical trials has become an important method. PDX clinical trials can be used for the development of anti-cancer drugs before clinical trials, with their efficacy assessed by the modified response evaluation criteria in solid tumors (mRECIST). A few dozen cases of PDX models have completed enrollment, and the efficacy of the drugs is assessed by 1 × 1 × 1 or 3 × 1 × 1 approaches in the PDX clinical trials. Furthermore, co-clinical trials can be used for personalized care or precision medicine with the evaluation of a new drug or a novel combination. Several PDX models from patients in clinical trials have been used to assess the efficacy of individual drugs or drug combinations in co-clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Clinical Trials as Topic , Humans , Neoplasms/pathology , Publications
15.
J Control Release ; 284: 49-56, 2018 08 28.
Article in English | MEDLINE | ID: mdl-29906553

ABSTRACT

Antibody-drug conjugates (ADCs) are currently considered to be promising agents for cancer therapy. However, especially in solid tumors, the uneven distribution of ADCs would decrease their efficacy in clinical studies. We suggest that in addition to optimizing ADC components, such as the linker structure and anticancer agent, it is necessary to consider the distribution of the ADC within tumor tissue. In this study, we established three kinds of anti-tissue factor (TF) ADCs: 1849ADC with a low kd, 444ADC with an intermediate kd, and 1084ADC with a high kd. All three of the anti-TF ADCs exhibited almost the same in vitro cytotoxicity and pharmacological and biochemical characteristics, although the binding kinetics parameters differed. In vivo, all ADCs exerted equivalent antitumor effects against small BxPC3 tumors. However, on larger BxPC3 tumors, 1084ADC (higher kd) exerted higher antitumor activity than 1849ADC (lower kd). Furthermore, immunofluorescence staining indicated that 1084ADC was distributed throughout the whole tumor, whereas 1849ADC was mainly localized close to tumor vessels. We conclude that the ADC with a higher kd increased the antitumor effect of because it penetrated and distributed evenly throughout the entire solid tumor. These findings highlight the importance of the kd of a mAb in ADC design.


Subject(s)
Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Pancreatic Neoplasms/drug therapy , Thromboplastin/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Female , Humans , Immunoconjugates/chemistry , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Thromboplastin/metabolism
16.
Bioengineering (Basel) ; 4(3)2017 Sep 17.
Article in English | MEDLINE | ID: mdl-28952557

ABSTRACT

Antibody-drug conjugate (ADC), as a next generation of antibody therapeutics, is a combination of an antibody and a drug connected via a specialized linker. ADC has four action steps: systemic circulation, the enhanced permeability and retention (EPR) effect, penetration within the tumor tissue, and action on cells, such as through drug delivery system (DDS) drugs. An antibody with a size of about 10 nm has the same capacity for passive targeting as some DDS carriers, depending on the EPR effect. In addition, some antibodies are capable of active targeting. A linker is stable in the bloodstream but should release drugs efficiently in the tumor cells or their microenvironment. Thus, the linker technology is actually a typical controlled release technology in DDS. Here, we focused on molecular imaging. Fluorescent and positron emission tomography (PET) imaging is useful for the visualization and evaluation of antibody delivery in terms of passive and active targeting in the systemic circulation and in tumors. To evaluate the controlled release of the ADC in the targeted area, a mass spectrometry imaging (MSI) with a mass microscope, to visualize the drug released from ADC, was used. As a result, we succeeded in confirming the significant anti-tumor activity of anti-fibrin, or anti-tissue factor-ADC, in preclinical settings by using DDS and molecular imaging.

17.
Sci Rep ; 7(1): 12341, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28951589

ABSTRACT

Nuclear medicine examinations for imaging gliomas have been introduced into clinical practice to evaluate the grade of malignancy and determine sampling locations for biopsies. However, these modalities have some limitations. Tissue factor (TF) is overexpressed in various types of cancers, including gliomas. We thus generated an anti-human TF monoclonal antibody (mAb) clone 1849. In the present study, immunohistochemistry performed on glioma specimens using anti-TF 1849 mAb showed that TF expression in gliomas increased in proportion to the grade of malignancy based on the World Health Organization (WHO) classification, and TF was remarkably expressed in necrosis and pseudopalisading cells, the histopathological hallmarks of glioblastoma multiforme (GBM). Furthermore, in both fluorescence and single-photon emission computed tomography/computed tomography (SPECT/CT) imaging studies, anti-TF 1849 IgG efficiently accumulated in TF-overexpressing intracranial tumours in mice. Although further investigation is required for a future clinical use of immuno-SPECT with 111In-labelled anti-TF 1849 IgG, the immuno-SPECT may represent a unique imaging modality that can visualize the biological characteristics of gliomas differently from those obtained using the existing imaging modalities and may be useful to evaluate the grade of malignancy and determine sampling locations for biopsies in patients with glioma, particularly GBM.


Subject(s)
Brain Neoplasms/diagnostic imaging , Glioma/diagnostic imaging , Immunoconjugates/administration & dosage , Molecular Imaging/methods , Thromboplastin/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Brain/diagnostic imaging , Brain/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Glioma/pathology , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Indium Radioisotopes/administration & dosage , Indium Radioisotopes/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Single Photon Emission Computed Tomography Computed Tomography , Thromboplastin/metabolism , Xenograft Model Antitumor Assays
18.
Yakugaku Zasshi ; 137(5): 535-544, 2017.
Article in Japanese | MEDLINE | ID: mdl-28458285

ABSTRACT

Antibody-drug conjugates (ADCs) comprise an antibody, a linker, and a drug or payload. The selection of a tumor-specific antibody and development of a linker having an efficient controlled drug release (CDR) are critical steps in developing a fully functional and effective ADC. In our research strategy, molecular imaging technologies have been employed to evaluate the efficiency of antibody delivery and CDR of the linker. In preclinical setting, antibody delivery into the tumor area or antibody penetration through the tumor stroma in malignant lymphoma or pancreatic tumor was evaluated by in vivo fluorescence imaging technique. Positron emission tomography (PET) imaging studies were conducted using 89Zr-labeled antibody to evaluate tumor targeting in a spontaneous carcinogenesis model. The model had dense stroma and was pathophysiologically very similar to human cancer. The drug imaging system, using microscopic mass spectroscopy (MMS) with enhanced resolution and sensitivity, was used for the evaluation of CDR. Paclitaxel (PTX)-incorporated micelle, a high-molecular-weight (HMW) carrier with CDR, showing similar properties as those of ADC, was analyzed. In contrast to free PTX, micelle selectively increased drug accumulation into the tumor and reduced toxicity in normal tissues by the enhanced permeability and retention (EPR) effect. Our drug imaging system has been used recently to evaluate the CDR of the ADC-linker. We present our work on the development of ADC using a molecular imaging technique.


Subject(s)
Antibodies, Monoclonal , Drug Delivery Systems , Drug Discovery/methods , Immunoconjugates , Molecular Imaging/methods , Humans , Mass Spectrometry , Microscopy , Molecular Targeted Therapy , Neoplasms/drug therapy
19.
Oncotarget ; 8(4): 6970-6983, 2017 Jan 24.
Article in English | MEDLINE | ID: mdl-28036302

ABSTRACT

INTRODUCTION: A standard treatment for stage II colorectal cancer (CRC) is surgical resection without adjuvant chemotherapy. However, the recurrence rate of these patients is approximately 20%. To date, there are no robust biomarkers suitable for predicting recurrence in stage II CRC patients. In this study, microRNAs (miRNAs) extracted from CRC tissues were examined for a possible biomarker to predict recurrence in stage II CRC patients. RESULTS: From the comprehensive analysis, 15 miRNAs were selected as candidates for further study. Regarding let-7a, -7d, -7e, miR-23c, -26b, -128a, -151-5p, and -181c, recurrence rates in training cohort patients with higher expression of these miRNAs isolated from their frozen tissues samples were significantly higher than those with lower expression (P < 0.05). According to multivariate analysis, the higher expression of miR-181c was detected as an independent predictive factor of recurrence (P = 0.001, OR: 9.43, 95% CI: 2.57-34.48). Results were similar in miR-181c extracted from FFPE tissues obtained from the training cohort (P = 0.003, OR: 7.46, 95% CI: 1.97-28.57). In the validation cohort using FFPE tissues, the recurrence rate in patients with higher miR-181c expression was significantly higher than those with lower miR-181c expression (P < 0.001). MATERIALS AND METHODS: Comprehensive analysis using a highly sensitive miRNA chip was initially performed to select candidate miRNAs associated with recurrence. Candidate miRNAs were analyzed by real-time RT-PCR using RNA from frozen and formalin-fixed, paraffin-embedded (FFPE) tissues. CONCLUSIONS: Higher expression of miR-181c may be a useful recurrence predictor of stage II CRC patients.


Subject(s)
Colorectal Neoplasms/pathology , MicroRNAs/genetics , Neoplasm Recurrence, Local/pathology , Up-Regulation , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Staging
20.
Oncotarget ; 8(7): 11020-11029, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28038457

ABSTRACT

Epidermal growth factor receptor (EGFR) is a well-validated oncological target molecule for monoclonal antibody therapies and Sym004 is a novel anti-EGFR antibody mixture comprising two recombinant chimeric IgG1 antibodies against non-overlapping epitopes of EGFR. Because EGFR is highly expressed in the majority of esophageal squamous cell carcinomas (ESCCs), we investigated the efficacy of Sym004 in human ESCC cell lines. Forty eight ESCC cell lines were treated with three kinds of anti-EGFR antibodies (Sym004, cetuximab, and panitumumab). Genetic background was investigated by next generation sequencing. The internalization of anti-EGFR antibodies into ESCC cells and inhibition of the EGFR signaling cascade by anti-EGFR antibodies were investigated in vitro. Furthermore, growth inhibition by anti-EGFR antibody treatment was investigated in vitro and in vivo. Sym004 treatments were more effective at inducing EGFR internalization and degradation than the two other anti-EGFR antibodies. Sym004 was more sensitive significantly to cell lines with EGFR gene amplification than those without amplification (P = 0.002). Growth inhibition of Sym004 was greater than in that of cetuximab or panitumumab in vitro and in vivo. These studies showed that Sym004 exhibited antitumor activity in some ESCC cell lines in preclinical settings and warrant a clinical evaluation in patients with ESCC. EGFR amplification is a potential biomarker of response to Sym004.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/drug therapy , ErbB Receptors/metabolism , Esophageal Neoplasms/drug therapy , Mice, Inbred BALB C , Xenograft Model Antitumor Assays/methods , Animals , Antibodies, Monoclonal/immunology , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cetuximab/pharmacology , ErbB Receptors/genetics , ErbB Receptors/immunology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Gene Amplification , Humans , Mice, Nude , Panitumumab , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...