151
|
Fu C, Zhu X, Xu P, Li Y. Pharmacological inhibition of USP7 promotes antitumor immunity and contributes to colon cancer therapy. Onco Targets Ther 2019; 12:609-617. [PMID: 30697058 PMCID: PMC6339463 DOI: 10.2147/ott.s182806] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Effectiveness of clinical therapy such as chemotherapy for solid tumors is limited by acquired drug resistance and side effects. Available antitumor immunity methods showed promising prospect of cancer therapy. However, more drug targets for boosting antitumor immunity still need to be explored and selective and effective compounds are yet to be developed. Purpose To study the effect and possible mechanism of compound P5091, a selective USP7 inhibitor, on CT26 xenografts growth in mice. Materials and methods CT26 xenografts model was employed to examine the anti-tumor effect of P5091. RT-PCR and ELISA analysis were used to detect the level of IFN-γ, TNF-α and IL-10 in tumor tissue and serum, respectively. IFN-γ expression in CD4+ and CD8+ T cells was analyzed by intracellular stain. The level of FOXP3 in Treg cells was confirmed by intracellular stain and western blotting. Results Compound P5091, a selective USP7 inhibitor, was found to inhibit CT26 xenografts growth in mice, which is comparable to the effect of Anti-PD-1 antibody. RT-PCR analysis showed that P5091 treatment decreased IL-10 mRNA level in tumor tissue while elevated mRNA level of IFN-γ and TNF-α. Moreover, ELISA analysis manifested decreased of IL-10 and elevation of IFN-γ and TNF-α in serum from tumor bearing mice. Intracellular stain showed increased IFN-g expression both in CD4+ and CD8+ T cells after P5091 treatment. Furthermore, P5091 treatment caused FOXP3 loss in Treg cells decreased the proportion of Treg cells in tumor bearing mice. Conclusion Our study here showed that P5091 may be a candidate for cancer immunotherapy.
Collapse
Affiliation(s)
- Changqing Fu
- Clinical Laboratory, Zhangjiagang Fifth People's Hospital, Suzhou University, Suzhou, Jiangsu 215621, People's Republic of China
| | - Xiaojue Zhu
- Clinical Laboratory, Zhangjiagang First People's Hospital, Suzhou University, Suzhou, Jiangsu 215600, People's Republic of China,
| | - Peiqi Xu
- Clinical Laboratory, Zhangjiagang First People's Hospital, Suzhou University, Suzhou, Jiangsu 215600, People's Republic of China,
| | - Yonghao Li
- Clinical Laboratory, Zhangjiagang First People's Hospital, Suzhou University, Suzhou, Jiangsu 215600, People's Republic of China,
| |
Collapse
|
152
|
Jian JW, Chen HS, Chiu YK, Peng HP, Tung CP, Chen IC, Yu CM, Tsou YL, Kuo WY, Hsu HJ, Yang AS. Effective binding to protein antigens by antibodies from antibody libraries designed with enhanced protein recognition propensities. MAbs 2019; 11:373-387. [PMID: 30526270 PMCID: PMC6380391 DOI: 10.1080/19420862.2018.1550320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Antibodies provide immune protection by recognizing antigens of diverse chemical properties, but elucidating the amino acid sequence-function relationships underlying the specificity and affinity of antibody-antigen interactions remains challenging. We designed and constructed phage-displayed synthetic antibody libraries with enriched protein antigen-recognition propensities calculated with machine learning predictors, which indicated that the designed single-chain variable fragment variants were encoded with enhanced distributions of complementarity-determining region (CDR) hot spot residues with high protein antigen recognition propensities in comparison with those in the human antibody germline sequences. Antibodies derived directly from the synthetic antibody libraries, without affinity maturation cycles comparable to those in in vivo immune systems, bound to the corresponding protein antigen through diverse conformational or linear epitopes with specificity and affinity comparable to those of the affinity-matured antibodies from in vivo immune systems. The results indicated that more densely populated CDR hot spot residues were sustainable by the antibody structural frameworks and could be accompanied by enhanced functionalities in recognizing protein antigens. Our study results suggest that synthetic antibody libraries, which are not limited by the sequences found in antibodies in nature, could be designed with the guidance of the computational machine learning algorithms that are programmed to predict interaction propensities to molecules of diverse chemical properties, leading to antibodies with optimal characteristics pertinent to their medical applications.
Collapse
Affiliation(s)
- Jhih-Wei Jian
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan.,b Institute of Biomedical Informatics, National Yang-Ming University , Taipei , Taiwan.,c Bioinformatics Program, Taiwan International Graduate Program , Institute of Information Science, Academia Sinica , Taipei , Taiwan
| | - Hong-Sen Chen
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Yi-Kai Chiu
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Hung-Pin Peng
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Chao-Ping Tung
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Ing-Chien Chen
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Chung-Ming Yu
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Yueh-Liang Tsou
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Wei-Ying Kuo
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - Hung-Ju Hsu
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| | - An-Suei Yang
- a Genomics Research Center , Academia Sinica , Taipei , Taiwan
| |
Collapse
|
153
|
Irwin DE, Davis B, Bell JA, Galaznik A, Garcia-Ribas I. Gastrointestinal complications in patients treated with ipilimumab and nivolumab combination therapy or monotherapy. J Comp Eff Res 2018; 8:81-90. [PMID: 30547675 DOI: 10.2217/cer-2018-0072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim & methods: A retrospective study using the IBM Explorys Universe Database assessed the risk of gastrointestinal events (enterocolitis or diarrhea) among melanoma and lung cancer patients treated with ipilimumab and nivolumab combination or monotherapy. Results & conclusion: There were 904 melanoma patients (607 ipilimumab, 140 nivolumab and 157 combo) and 1641 lung cancer patients (68 ipilimumab, 1542 nivolumab and 31 combo). Approximately, 37% of lung patients and 46% of melanoma patients experienced at least one adverse event. After adjusting for covariates, patients receiving combination therapy were more likely to have a gastrointestinal event compared with ipilimumab monotherapy patients (melanoma hazard ratio: 1.54; 95% CI: 1.06-2.25; lung hazard ratio: 2.93; 95% CI: 1.09-7.89).
Collapse
Affiliation(s)
- Debra E Irwin
- Truven Health Analytics, an IBM Company, 75 Binney Street, Cambridge, MA 02142, USA
| | - Brian Davis
- Truven Health Analytics, an IBM Company, 75 Binney Street, Cambridge, MA 02142, USA
| | - Jill A Bell
- Takeda Oncology, a wholly owned subsidiary of Takeda Pharmaceutical Company, Limited, 40 Landsdowne Street, Cambridge, MA 02139, USA
| | - Aaron Galaznik
- Takeda Oncology, a wholly owned subsidiary of Takeda Pharmaceutical Company, Limited, 40 Landsdowne Street, Cambridge, MA 02139, USA
| | - Ignacio Garcia-Ribas
- Takeda Oncology, a wholly owned subsidiary of Takeda Pharmaceutical Company, Limited, 40 Landsdowne Street, Cambridge, MA 02139, USA
| |
Collapse
|
154
|
Xie K, Song S, Zhou L, Wan J, Qiao Y, Wang M, Xie H, Zhou L, Zheng S, Wang H. Revival of a potent therapeutic maytansinoid agent using a strategy that combines covalent drug conjugation with sequential nanoparticle assembly. Int J Pharm 2018; 556:159-171. [PMID: 30553007 DOI: 10.1016/j.ijpharm.2018.12.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 11/10/2018] [Accepted: 12/03/2018] [Indexed: 01/16/2023]
Abstract
Maytansine and its related analogues are a class of highly potent anti-proliferation agents that have failed to be exploited as clinical drugs for human therapy due to unacceptable systemic toxicity. Here, we delineate a novel strategy that combines rational drug conjugation with subsequent nanoparticle assembly to systemically deliver this highly potent and toxic drug. To demonstrate this concept, we covalently coupled the thiolated maytansine derivative, the DM1 agent, to amphiphilic block co-polymers, polyethylene glycol (PEG)-block-polylactide (PLA), in varying molecular weights to generate two prodrug constructs (i.e., PEG2K-PLA2K-DM1 and PEG2K-PLA4K-DM1) via the maleimide-thiol reaction. The resulting two constructs are amenable to self-assembly in aqueous solutions and are systemically injectable for preclinical studies. In vivo evaluations indicate that PEG-PLA-DM1 conjugate-assembled nanoparticles (NPs) display substantially reduced drug toxicity compared to the free drug forms and NPs that physically encapsulate DM1. Furthermore, following systemic administration, these nanodrugs produced superior therapeutic efficacy over free DM1 in a colon tumor xenograft-bearing mouse model. Therefore, this study provides evidence that the conjugation of toxic drugs to assembling copolymers enables the alleviation of cancer drug toxicity and effective delivery of anticancer drugs. Thus, this DM1-formulated platform represents a new generation of nanotherapeutics that are available for further clinical evaluation.
Collapse
Affiliation(s)
- Ke Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Shanshan Song
- Institute of Microanalytical Systems, Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China
| | - Liqian Zhou
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Jianqin Wan
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Yiting Qiao
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Min Wang
- Institute of Microanalytical Systems, Department of Chemistry, Zhejiang University, Hangzhou 310058, PR China
| | - Haiyang Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Lin Zhou
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Shusen Zheng
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China.
| | - Hangxiang Wang
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou 310003, PR China.
| |
Collapse
|
155
|
Murer P, Kiefer JD, Plüss L, Matasci M, Blümich SL, Stringhini M, Neri D. Targeted Delivery of TNF Potentiates the Antibody-Dependent Cell-Mediated Cytotoxicity of an Anti-Melanoma Immunoglobulin. J Invest Dermatol 2018; 139:1339-1348. [PMID: 30543899 DOI: 10.1016/j.jid.2018.11.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/01/2018] [Accepted: 11/13/2018] [Indexed: 12/27/2022]
Abstract
The recombinant murine IgG2a antibody TA99, directed against a melanoma antigen, was used to study combination modalities that potentiate antibody-dependent cell cytotoxicity. As previously reported, IgG2a(TA99) was extremely efficacious in preventing the growth of B16 lung metastases. However, the same antibody mediated only minimal tumor growth retardation when used to treat established neoplastic masses. The therapeutic activity of IgG2a(TA99) could be substantially enhanced by co-administration with an antibody-cytokine fusion (TA99-murine tumor necrosis factor [mTNF]), consisting of the TA99 antibody in single-chain variable fragment format fused to murine TNF. This fusion protein efficiently killed endothelial cells in vitro and displayed only minimal activity against B16 melanoma cells. In vivo, TA99-mTNF boosted the influx of natural killer cells and macrophages into B16 melanoma lesions. Therapy studies with two different administration schedules showed that the combination of TA99-mTNF and IgG2a(TA99) was superior to the individual products used as single agents. The combination treatment converted most of the tumor mass into a necrotic lesion, but a vital tumor rim eventually regrew, even when dacarbazine was included in the therapeutic regimen. The treatment modality described in this article may be applicable to the treatment of melanoma patients, given the specificity of the gp75 antigen and its conservation across species.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/administration & dosage
- Antibodies, Monoclonal, Murine-Derived/genetics
- Antibodies, Monoclonal, Murine-Derived/isolation & purification
- Antigens, Neoplasm/immunology
- CHO Cells
- Cell Line, Tumor/transplantation
- Cricetulus
- Drug Administration Schedule
- Drug Screening Assays, Antitumor
- Female
- Humans
- Immunoconjugates/administration & dosage
- Immunoconjugates/genetics
- Immunoconjugates/isolation & purification
- Immunoglobulin G/immunology
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Membrane Glycoproteins/immunology
- Mice
- Oxidoreductases/immunology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/isolation & purification
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Tumor Necrosis Factor-alpha/administration & dosage
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/isolation & purification
Collapse
Affiliation(s)
- Patrizia Murer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Jonathan D Kiefer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Louis Plüss
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | | | - Sandra L Blümich
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, University of Zurich, Zurich, Switzerland
| | - Marco Stringhini
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland.
| |
Collapse
|
156
|
Poot AJ, Adamzek KWA, Windhorst AD, Vosjan MJWD, Kropf S, Wester HJ, van Dongen GAMS, Vugts DJ. Fully Automated 89Zr Labeling and Purification of Antibodies. J Nucl Med 2018; 60:691-695. [PMID: 30530830 DOI: 10.2967/jnumed.118.217158] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/24/2018] [Indexed: 01/05/2023] Open
Abstract
Dozens of monoclonal antibodies (mAbs) have been approved for clinical use, and hundreds more are under development. To support these developments and facilitate a personalized medicine approach, PET imaging and quantification of mAbs, after chelation with desferrioxamine B (DFO) and radiolabeling with 89Zr, has become attractive. Also, the use of 89Zr-mAbs in preclinical and clinical studies is expanding rapidly. Despite these rapid developments, 89Zr radiolabeling is still performed manually. Therefore, we aimed to develop a simple, fully automated, good-manufacturing-practice (GMP)-compliant production procedure for the 89Zr labeling of mAbs. Such procedures should increase the robustness and capacity of 89Zr-mAb production while minimizing the radiation dose to the operator. Here, the procedures for fully automated 89Zr-mAb production are described and applied to produce batches of 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab suitable for clinical use. Both products had to meet the GMP-compliant quality standards with respect to yield, radiochemical purity, protein integrity, antigen binding, sterility, and endotoxin levels. Methods: Automated 89Zr labeling of mAbs was developed on a Scintomics GRP 2V module and comprised the following steps: reagent transfer to the 89Zr-containing reaction vial, mixing of the reagents followed by a 60-min reaction at room temperature to obtain optimal radiolabeling yields, and product purification using a PD-10 desalting column. Results: Radiochemical yields of 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab were all more than 90% according to instant thin-layer chromatography. Isolated yields were 74.6% ± 2.0% and 62.6% ± 3.0% for 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab, respectively, which are similar to isolated yields obtained using GMP protocols for manual 89Zr labeling of mAbs. To meet the GMP-compliant quality standards, only the radiochemically pure fractions were collected from PD-10, resulting in a lower isolated yield than the radiochemical yield according to instant thin-layer chromatography. The radiochemical purity and protein integrity were more than 95% for both products, and the antigen binding was 95.6% ± 0.6% and 87.1% ± 2.2% for 89Zr-DFO-N-suc-cetuximab and 89Zr-DFO-N-suc-rituximab, respectively. The products were sterile, and the endotoxin levels were within acceptable limits, allowing future clinical production using this procedure. Conclusion: Procedures for fully automated GMP-compliant production of 89Zr-mAbs were developed on a commercially available synthesis module, which also allows the GMP production of other radiolabeled mAbs.
Collapse
Affiliation(s)
- Alex J Poot
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Kevin W A Adamzek
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Saskia Kropf
- Scintomics GmbH, Fuerstenfeldbruck, Germany; and
| | - Hans-Jurgen Wester
- Scintomics GmbH, Fuerstenfeldbruck, Germany; and.,Pharmaceutical Radiochemistry, Technische Universität München, Garching, Germany
| | - Guus A M S van Dongen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Danielle J Vugts
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
157
|
Weimbs T, Shillingford JM, Torres J, Kruger SL, Bourgeois BC. Emerging targeted strategies for the treatment of autosomal dominant polycystic kidney disease. Clin Kidney J 2018; 11:i27-i38. [PMID: 30581563 PMCID: PMC6295603 DOI: 10.1093/ckj/sfy089] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/27/2018] [Indexed: 12/25/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a widespread genetic disease that leads to renal failure in the majority of patients. The very first pharmacological treatment, tolvaptan, received Food and Drug Administration approval in 2018 after previous approval in Europe and other countries. However, tolvaptan is moderately effective and may negatively impact a patient's quality of life due to potentially significant side effects. Additional and improved therapies are still urgently needed, and several clinical trials are underway, which are discussed in the companion paper Müller and Benzing (Management of autosomal-dominant polycystic kidney disease-state-of-the-art) Clin Kidney J 2018; 11: i2-i13. Here, we discuss new therapeutic avenues that are currently being investigated at the preclinical stage. We focus on mammalian target of rapamycin and dual kinase inhibitors, compounds that target inflammation and histone deacetylases, RNA-targeted therapeutic strategies, glucosylceramide synthase inhibitors, compounds that affect the metabolism of renal cysts and dietary restriction. We discuss tissue targeting to renal cysts of small molecules via the folate receptor, and of monoclonal antibodies via the polymeric immunoglobulin receptor. A general problem with potential pharmacological approaches is that the many molecular targets that have been implicated in ADPKD are all widely expressed and carry out important functions in many organs and tissues. Because ADPKD is a slowly progressing, chronic disease, it is likely that any therapy will have to continue over years and decades. Therefore, systemically distributed drugs are likely to lead to potentially prohibitive extra-renal side effects during extended treatment. Tissue targeting to renal cysts of such drugs is one potential way around this problem. The use of dietary, instead of pharmacological, interventions is another.
Collapse
Affiliation(s)
- Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Jonathan M Shillingford
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Jacob Torres
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Samantha L Kruger
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Bryan C Bourgeois
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| |
Collapse
|
158
|
Jiao H, Zhao X, Liu J, Ma T, Zhang Z, Zhang J, Wang J. In vivo imaging characterization and anticancer efficacy of a novel HER2 affibody and pemetrexed conjugate in lung cancer model. Nucl Med Biol 2018; 68-69:31-39. [PMID: 30578135 DOI: 10.1016/j.nucmedbio.2018.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/05/2018] [Accepted: 11/22/2018] [Indexed: 11/26/2022]
Abstract
INTRODUCTION In this study, a new agent consisting of HER2-specific affibody ZHER2:V2 and chemotherapy drug pemetrexed was synthesized to develop a new targeted drug. Its biological characteristics and anticancer efficacy were assessed in cells level and xenografts models by radiolabeling with technetium-99m. METHODS After the ZHER2:V2-pemetrexed conjugate was synthesized, radiolabeling of the conjugate was performed using its C-terminal 4 amino acids (Gly-Gly-Gly-Cys) as the chelating moiety. The radiochemical yield of the [99mTc]Tc-ZHER2:V2-pemetrexed was identified by instant thin-layer chromatography (ITLC). Stability of the radiolabeled conjugate was investigated both in vitro and in vivo. In vitro binding affinity and cell internalization study of the probe were performed in A549 cells (HER2-positive). Tumor uptake was evaluated by in vitro uptake assay in A549 cells and H23 cells (HER2-negative), and by in vivo biodistribution and SPECT imaging in A549 and H23 tumor-bearing mice. The antitumor efficacy of the ZHER2:V2-pemetrexed conjugate was evaluated in cells and xenograft models. RESULTS The ZHER2:V2-pemetrexed was successfully synthesized and conjugated with technetium-99 m, and acquired the radiochemical yield of 97.0 ± 0.3%. The stability of [99mTc]Tc-ZHER2:V2-pemetrexed was good in both physiological saline and human serum. The radiolabeled agent displayed excellent HER2-binding specificity and affinity in vitro, and was gradually internalized into the cells. Biodistribution study revealed obvious tumor uptake in A549 xenografts (percentage injected dose per gram, 2.6 ± 1.0%ID/g at 4 h postinjection), while the uptake in HER2-negative H23 tumors was much lower (0.2 ± 0.1%ID/g at 4 h postinjection, P < 0.01). SPECT imaging exhibited an intensity in the A549 xenograft which could be blocked by excess ZHER2:V2-pemetrexed. Treatment with ZHER2:V2-pemetrexed significantly impaired the tumor growth (P < 0.05), with less weight loss than pemetrexed. CONCLUSION [99mTc]Tc-ZHER2:V2-pemetrexed showed desirable property and HER2-specificity. The ZHER2:V2-pemetrexed conjugate could inhibit tumor growth of HER2-positive lung adenocarcinoma and may have the potential to become a targeted drug for lung cancer. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE The compound described herein performs HER2-targeting with favorable anticancer efficacy and offers the potential of novel targeting strategies for further tumor therapy.
Collapse
Affiliation(s)
- Honglei Jiao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Jiahui Liu
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Tuo Ma
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
159
|
Jiang Y, Zhang J, Meng F, Zhong Z. Apolipoprotein E Peptide-Directed Chimeric Polymersomes Mediate an Ultrahigh-Efficiency Targeted Protein Therapy for Glioblastoma. ACS NANO 2018; 12:11070-11079. [PMID: 30395440 DOI: 10.1021/acsnano.8b05265] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The inability to cross the blood-brain barrier (BBB) prevents nearly all chemotherapeutics and biotherapeutics from the effective treatment of brain tumors, rendering few improvements in patient survival rates to date. Here, we report that apolipoprotein E peptide [ApoE, (LRKLRKRLL)2C] specifically binds to low-density lipoprotein receptor members (LDLRs) and mediates superb BBB crossing and highly efficient glioblastoma (GBM)-targeted protein therapy in vivo. The in vitro BBB model studies reveal that ApoE induces 2.2-fold better penetration of the immortalized mouse brain endothelial cell line (bEnd.3) monolayer for chimeric polymersomes (CP) compared to Angiopep-2, the best-known BBB-crossing peptide used in clinical trials for GBM therapy. ApoE-installed CP (ApoE-CP) carrying saporin (SAP) displays a highly specific and potent antitumor effect toward U-87 MG cells with a low half-maximum inhibitory concentration of 14.2 nM SAP. Notably, ApoE-CP shows efficient BBB crossing as well as accumulation and penetration in orthotopic U-87 MG glioblastoma. The systemic administration of SAP-loaded ApoE-CP causes complete growth inhibition of orthotopic U-87 MG GBM without eliciting any observable adverse effects, affording markedly improved survival benefits. ApoE peptide provides an ultrahigh-efficiency targeting strategy for GBM therapy.
Collapse
Affiliation(s)
- Yu Jiang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , PR China
| | - Jian Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection , Soochow University , Suzhou 215123 , PR China
| |
Collapse
|
160
|
Affiliation(s)
- Xun Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Fan Wu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| | - Yong Ji
- Department of Cardiothoracic Surgery, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Lichen Yin
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou 215123, China
| |
Collapse
|
161
|
Benonisson H, Sow HS, Breukel C, Claassens J, Brouwers C, Linssen MM, Fransen MF, Sluijter M, Ossendorp F, van Hall T, Verbeek JS. High FcγR Expression on Intratumoral Macrophages Enhances Tumor-Targeting Antibody Therapy. THE JOURNAL OF IMMUNOLOGY 2018; 201:3741-3749. [PMID: 30397036 DOI: 10.4049/jimmunol.1800700] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022]
Abstract
Therapy with tumor-specific Abs is common in the clinic but has limited success against solid malignancies. We aimed at improving the efficacy of this therapy by combining a tumor-specific Ab with immune-activating compounds. In this study, we demonstrate in the aggressive B16F10 mouse melanoma model that concomitant application of the anti-TRP1 Ab (clone TA99) with TLR3-7/8 or -9 ligands, and IL-2 strongly enhanced tumor control in a therapeutic setting. Depletion of NK cells, macrophages, or CD8+ T cells all mitigated the therapeutic response, showing a coordinated immune rejection by innate and adaptive immune cells. FcγRs were essential for the therapeutic effect, with a dominant role for FcγRI and a minor role for FcγRIII and FcγRIV. FcγR expression on NK cells and granulocytes was dispensable, indicating that other tumoricidal functions of NK cells were involved and implicating that FcγRI, -III, and -IV exerted their activity on macrophages. Indeed, F4/80+Ly-6C+ inflammatory macrophages in the tumor microenvironment displayed high levels of these receptors. Whereas administration of the anti-TRP1 Ab alone reduced the frequency of these macrophages, the combination with a TLR agonist retained these cells in the tumor microenvironment. Thus, the addition of innate stimulatory compounds, such as TLR ligands, to tumor-specific Ab therapy could greatly enhance its efficacy in solid cancers via optimal exploitation of FcγRs.
Collapse
Affiliation(s)
- Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Heng Sheng Sow
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Cor Breukel
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jill Claassens
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Conny Brouwers
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Margot M Linssen
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands;
| |
Collapse
|
162
|
A Two-Step Approach for the Design and Generation of Nanobodies. Int J Mol Sci 2018; 19:ijms19113444. [PMID: 30400198 PMCID: PMC6274671 DOI: 10.3390/ijms19113444] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/29/2022] Open
Abstract
Nanobodies, the smallest possible antibody format, have become of considerable interest for biotechnological and immunotherapeutic applications. They show excellent robustness, are non-immunogenic in humans, and can easily be engineered and produced in prokaryotic hosts. Traditionally, nanobodies are selected from camelid immune libraries involving the maintenance and treatment of animals. Recent advances have involved the generation of nanobodies from naïve or synthetic libraries. However, such approaches demand large library sizes and sophisticated selection procedures. Here, we propose an alternative, two-step approach for the design and generation of nanobodies. In a first step, complementarity-determining regions (CDRs) are grafted from conventional antibody formats onto nanobody frameworks, generating weak antigen binders. In a second step, the weak binders serve as templates to design focused synthetic phage libraries for affinity maturation. We validated this approach by grafting toxin- and hapten-specific CDRs onto frameworks derived from variable domains of camelid heavy-chain-only antibodies (VHH). We then affinity matured the hapten binder via panning of a synthetic phage library. We suggest that this strategy can complement existing immune, naïve, and synthetic library based methods, requiring neither animal experiments, nor large libraries, nor sophisticated selection protocols.
Collapse
|
163
|
Payandeh Z, Noori E, Khalesi B, Mard-Soltani M, Abdolalizadeh J, Khalili S. Anti-CD37 targeted immunotherapy of B-Cell malignancies. Biotechnol Lett 2018; 40:1459-1466. [PMID: 30293139 DOI: 10.1007/s10529-018-2612-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022]
Abstract
CD37 is a member of tetra-spanning superfamily (characterized by their four transmembrane domains). It is one of the specific proteins for normal and malignant mature B cells. Anti CD37 monoclonal antibodies are reported to improve the overall survival in CLL. These therapeutics will increase the efficacy and reduce the toxicity in patients with both newly diagnosed and relapsed and refractory disease. Recent clinical trials have shown promising outcomes for these agents, administered both as monotherapy and in combination with standard chemotherapeutics. Long-term follow-up of combination regimens has even raised the question of whether the patients with CLL could be treated with intensive chemo-immunotherapy. In the present study, CD37 is introduced as an appealing target to treat B cell malignancies. The anti-CD37 antibodies as one of the most successful therapeutics against CD37 are introduced and the clinical outcomes of their exploitation are explained.
Collapse
Affiliation(s)
- Zahra Payandeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Effat Noori
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Maysam Mard-Soltani
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| |
Collapse
|
164
|
Yao P, Zhang Y, Meng H, Sun H, Zhong Z. Smart Polymersomes Dually Functionalized with cRGD and Fusogenic GALA Peptides Enable Specific and High-Efficiency Cytosolic Delivery of Apoptotic Proteins. Biomacromolecules 2018; 20:184-191. [DOI: 10.1021/acs.biomac.8b01243] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Peili Yao
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Hao Meng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Huanli Sun
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
165
|
An Immunoscore Using PD-L1, CD68, and Tumor-infiltrating Lymphocytes (TILs) to Predict Response to Neoadjuvant Chemotherapy in Invasive Breast Cancer. Appl Immunohistochem Mol Morphol 2018; 26:611-619. [DOI: 10.1097/pai.0000000000000485] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
166
|
Cui J, Björnmalm M, Ju Y, Caruso F. Nanoengineering of Poly(ethylene glycol) Particles for Stealth and Targeting. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:10817-10827. [PMID: 30132674 DOI: 10.1021/acs.langmuir.8b02117] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The assembly of particles composed solely or mainly of poly(ethylene glycol) (PEG) is an emerging area that is gaining increasing interest within bio-nano science. PEG, widely considered to be the "gold standard" among polymers for drug delivery, is providing a platform for exploring fundamental questions and phenomena at the interface between particle engineering and biomedicine. These include the targeting and stealth behaviors of synthetic nanomaterials in biological environments. In this feature article, we discuss recent work in the nanoengineering of PEG particles and explore how they are enabling improved targeting and stealth performance. Specific examples include PEG particles prepared through surface-initiated polymerization, mesoporous silica replication via postinfiltration, and particle assembly through metal-phenolic coordination. This particle class exhibits unique in vivo behavior (e.g., biodistribution and immune cell interactions) and has recently been explored for drug delivery applications.
Collapse
Affiliation(s)
- Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, and the School of Chemistry and Chemical Engineering , Shandong University , Jinan , Shandong 250100 , China
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
| | - Mattias Björnmalm
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering , Imperial College London , London SW7 2AZ , United Kingdom
| | - Yi Ju
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and the Department of Chemical Engineering , The University of Melbourne , Parkville , Victoria 3010 , Australia
| |
Collapse
|
167
|
Nanomedicines for developing cancer nanotherapeutics: from benchtop to bedside and beyond. Appl Microbiol Biotechnol 2018; 102:9449-9470. [DOI: 10.1007/s00253-018-9352-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/29/2018] [Accepted: 08/29/2018] [Indexed: 12/21/2022]
|
168
|
Nicolaou KC, Li R, Lu Z, Pitsinos EN, Alemany LB, Aujay M, Lee C, Sandoval J, Gavrilyuk J. Streamlined Total Synthesis of Shishijimicin A and Its Application to the Design, Synthesis, and Biological Evaluation of Analogues thereof and Practical Syntheses of PhthNSSMe and Related Sulfenylating Reagents. J Am Chem Soc 2018; 140:12120-12136. [PMID: 30216054 DOI: 10.1021/jacs.8b06955] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Shishijimicin A is a scarce marine natural product with highly potent cytotoxicities, making it a potential payload or a lead compound for designed antibody-drug conjugates. Herein, we describe an improved total synthesis of shishijimicin A and the design, synthesis, and biological evaluation of a series of analogues. Equipped with appropriate functionalities for linker attachment, a number of these analogues exhibited extremely potent cytotoxicities for the intended purposes. The synthetic strategies and tactics developed and employed in these studies included improved preparation of previously known and new sulfenylating reagents such as PhthNSSMe and related compounds.
Collapse
Affiliation(s)
| | | | | | - Emmanuel N Pitsinos
- Laboratory of Natural Products Synthesis & Bioorganic Chemistry, Institute of Nanoscience and Nanotechnology , National Centre for Scientific Research "Demokritos" , 153 10 Agia Paraskevi , Greece
| | | | - Monette Aujay
- AbbVie Stemcentrx, LLC , 450 East Jamie Court , South San Francisco , California 94080 , United States
| | - Christina Lee
- AbbVie Stemcentrx, LLC , 450 East Jamie Court , South San Francisco , California 94080 , United States
| | - Joseph Sandoval
- AbbVie Stemcentrx, LLC , 450 East Jamie Court , South San Francisco , California 94080 , United States
| | - Julia Gavrilyuk
- AbbVie Stemcentrx, LLC , 450 East Jamie Court , South San Francisco , California 94080 , United States
| |
Collapse
|
169
|
Xu Z, Park Y, Zhen B, Zhu B. Designing cancer immunotherapy trials with random treatment time-lag effect. Stat Med 2018; 37:4589-4609. [PMID: 30203592 DOI: 10.1002/sim.7937] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 11/07/2022]
Abstract
In some clinical settings such as the cancer immunotherapy trials, a treatment time-lag effect may be present and the lag duration possibly vary from subject to subject. An efficient study design and analysis procedure should not only take into account the time-lag effect but also consider the individual heterogeneity in the lag duration. In this paper, we present a Generalized Piecewise Weighted Logrank (GPW-Logrank) test, designed to account for the random time-lag effect while maximizing the study power with respect to the weights. Based on the proposed test, both analytic and numeric approaches are developed for the sample size and power calculation. Asymptotic properties are derived and finite sample efficiency is evaluated in simulations. Compared with the standard practice ignoring the delayed effect, the proposed design and analysis procedures are substantially more efficient when a random lag is expected; further, compared with the existing methods by Xu et al considering the fixed time-lag effect, the proposed approaches are significantly more robust when the lag model is misspecified. An R package (DelayedEffect.Design) is developed for implementation.
Collapse
Affiliation(s)
- Zhenzhen Xu
- CBER, Food and Drug Administration, Silver Spring, Maryland
| | - Yongsoek Park
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Boguang Zhen
- CBER, Food and Drug Administration, Silver Spring, Maryland
| | - Bin Zhu
- DCEG, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
170
|
Khoshtinat Nikkhoi S, Rahbarizadeh F, Ahmadvand D, Moghimi SM. Multivalent targeting and killing of HER2 overexpressing breast carcinoma cells with methotrexate-encapsulated tetra-specific non-overlapping variable domain heavy chain anti-HER2 antibody-PEG-liposomes: In vitro proof-of-concept. Eur J Pharm Sci 2018; 122:42-50. [DOI: 10.1016/j.ejps.2018.06.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 06/09/2018] [Accepted: 06/18/2018] [Indexed: 12/17/2022]
|
171
|
Yang D, Han Z, Alam MM, Oppenheim JJ. High-mobility group nucleosome binding domain 1 (HMGN1) functions as a Th1-polarizing alarmin. Semin Immunol 2018; 38:49-53. [PMID: 29503123 DOI: 10.1016/j.smim.2018.02.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/26/2018] [Indexed: 12/16/2022]
Abstract
High-mobility group (HMG) nucleosome binding domain 1 (HMGN1), which previously was thought to function only as a nucleosome-binding protein that regulates chromatin structure, histone modifications, and gene expression, was recently discovered to be an alarmin that contributes extracellularly to the generation of innate and adaptive immune responses. HMGN1 promotes DC recruitment through interacting with a Gαi protein-coupled receptor (GiPCR) and activates DCs predominantly through triggering TLR4. HMGN1 preferentially promotes Th1-type immunity, which makes it relevant for the fields of vaccinology, autoimmunity, and oncoimmunology. Here, we discuss the alarmin properties of HMGN1 and update recent advances on its roles in immunity and potential applications for immunotherapy of tumors.
Collapse
Affiliation(s)
- De Yang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA.
| | - Zhen Han
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA
| | - Md Masud Alam
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA
| | - Joost J Oppenheim
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, National Institute of Health, USA.
| |
Collapse
|
172
|
Mills CE, Michaud Z, Olsen BD. Elastin-like Polypeptide (ELP) Charge Influences Self-Assembly of ELP-mCherry Fusion Proteins. Biomacromolecules 2018; 19:2517-2525. [PMID: 29791150 DOI: 10.1021/acs.biomac.8b00147] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Self-assembly of protein-polymer bioconjugates presents an elegant strategy for controlling nanostructure and orientation of globular proteins in functional materials. Recent work has shown that genetic fusion of globular protein mCherry to an elastin-like polypeptide (ELP) yields similar self-assembly behavior to these protein-polymer bioconjugates. In the context of studying protein-polymer bioconjugate self-assembly, the mutability of the ELP sequence allows several different properties of the ELP block to be tuned orthogonally while maintaining consistent polypeptide backbone chemistry. This work uses this ELP sequence tunability in combination with the precise control offered by genetic engineering of an amino acid sequence to generate a library of four novel ELP sequences that are used to study the combined effect of charge and hydrophobicity on ELP-mCherry fusion protein self-assembly. Concentrated solution self-assembly is studied by small-angle X-ray scattering (SAXS) and depolarized light scattering (DPLS). These experiments show that fusions containing a negatively charged ELP block do not assemble at all, and fusions with a charge balanced ELP block exhibit a weak propensity for assembly. By comparison, the fusion containing an uncharged ELP block starts to order at 40 wt % in solution and at all concentrations measured has sharper, more intense SAXS peaks than other fusion proteins. These experiments show that charge character of the ELP block is a stronger predictor of self-assembly behavior than the hydrophobicity of the ELP block. Dilute solution small-angle neutron scattering (SANS) on the ELPs alone suggests that all ELPs used in this study (including the uncharged ELP) adopt dilute solution conformations similar to those of traditional polymers, including polyampholytes and polyelectrolytes. Finally, dynamic light scattering studies on ELP-mCherry blends shows that there is no significant complexation between the charged ELPs and mCherry. Therefore, it is proposed that the superior self-assembly of fusion proteins containing uncharged ELP block is due to effective repulsions between charged and uncharged blocks due to local charge correlation effects and, in the case of anionic ELPs, repulsion between like charges within the ELP block.
Collapse
Affiliation(s)
- Carolyn E Mills
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Zachary Michaud
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Bradley D Olsen
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
173
|
Fu W, Sun H, Zhao Y, Chen M, Yang L, Yang X, Jin W. Targeted delivery of CD44s-siRNA by ScFv overcomes de novo resistance to cetuximab in triple negative breast cancer. Mol Immunol 2018; 99:124-133. [DOI: 10.1016/j.molimm.2018.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/28/2018] [Accepted: 05/10/2018] [Indexed: 01/02/2023]
|
174
|
|
175
|
Matsuda A, Mimura M, Maruyama T, Kurinomaru T, Shiuhei M, Shiraki K. Liquid Droplet of Protein-Polyelectrolyte Complex for High-Concentration Formulations. J Pharm Sci 2018; 107:2713-2719. [PMID: 29960025 DOI: 10.1016/j.xphs.2018.06.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/22/2018] [Accepted: 06/20/2018] [Indexed: 11/26/2022]
Abstract
The formulation of high-concentration protein solutions is a challenging issue for achieving subcutaneous administration. Previously, we developed a method of precipitation-redissolution using polyelectrolyte as a precipitant to produce protein solutions at high concentrations. However, the redissolution yield of proteins was insufficient. This study aims to optimize the solution conditions for practical applications by combining IgG and poly-l-(glutamic acid) (polyE). A systematic analysis of solution pH and polyE size conditions revealed that an acidic condition favors precipitation, whereas neutral pH values are more effective for the redissolution. We find that the optimal size for polyE ranged from 15,000 to 50,000. This slight modification in the procedure in comparison with previous studies increased the precipitation and redissolution yields to nearly 100%, without irreversible protein denaturation. The fully reversible IgG-polyE complex formed as a droplet structure, which is similar to a condensate of liquid-liquid phase separation. The droplet structure plays an indispensable role in the salt-induced, redissolved state, which is pertinent to the new application that takes advantage of the methods to produce highly concentrated protein solutions.
Collapse
Affiliation(s)
- Ayumi Matsuda
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| | - Masahiro Mimura
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| | - Takuya Maruyama
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan
| | - Takaaki Kurinomaru
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Mieda Shiuhei
- Research and Development Center, Terumo Corporation, Nakai-machi, Ashigarakami-gun, Kanagawa 259-0151, Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan.
| |
Collapse
|
176
|
Watanabe Y, Vasiljevic S, Allen JD, Seabright GE, Duyvesteyn HME, Doores KJ, Crispin M, Struwe WB. Signature of Antibody Domain Exchange by Native Mass Spectrometry and Collision-Induced Unfolding. Anal Chem 2018; 90:7325-7331. [PMID: 29757629 PMCID: PMC6008249 DOI: 10.1021/acs.analchem.8b00573] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of domain-exchanged antibodies offers a route to high-affinity targeting to clustered multivalent epitopes, such as those associated with viral infections and many cancers. One strategy to generate these antibodies is to introduce mutations into target antibodies to drive domain exchange using the only known naturally occurring domain-exchanged anti-HIV (anti-human immunodeficiency virus) IgG1 antibody, 2G12 , as a template. Here, we show that domain exchange can be sensitively monitored by ion-mobility mass spectrometry and gas-phase collision-induced unfolding. Using native 2G12 and a mutated form that disrupts domain exchange such that it has a canonical IgG1 architecture ( 2G12 I19R ), we show that the two forms can be readily distinguished by their unfolding profiles. Importantly, the same signature of domain exchange is observed for both intact antibody and isolated Fab fragments. The development of a mass spectrometric method to detect antibody domain exchange will enable rapid screening and selection of candidate antibodies engineered to exhibit this and other unusual quaternary antibody architectures.
Collapse
Affiliation(s)
- Yasunori Watanabe
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, OX1 3QU, United Kingdom
- Biological Sciences & the Institute for Life Sciences, University of Southampton, SO17 1BJ, United Kingdom
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Roosevelt Drive, OX3 7BN, United Kingdom
| | - Snezana Vasiljevic
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, OX1 3QU, United Kingdom
| | - Joel D. Allen
- Biological Sciences & the Institute for Life Sciences, University of Southampton, SO17 1BJ, United Kingdom
| | - Gemma E. Seabright
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, OX1 3QU, United Kingdom
- Biological Sciences & the Institute for Life Sciences, University of Southampton, SO17 1BJ, United Kingdom
| | - Helen M. E. Duyvesteyn
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Roosevelt Drive, OX3 7BN, United Kingdom
| | - Katie J. Doores
- Department of Infectious Diseases, King’s College London, SE1 9RT, United Kingdom
| | - Max Crispin
- Biological Sciences & the Institute for Life Sciences, University of Southampton, SO17 1BJ, United Kingdom
| | - Weston B. Struwe
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, OX1 3QU, United Kingdom
- Chemistry Research Laboratory, Department of Chemistry, University of Oxford, OX1 3QZ, United Kingdom
| |
Collapse
|
177
|
Lee BC, Chalouni C, Doll S, Nalle SC, Darwish M, Tsai SP, Kozak KR, Del-Rosario G, Yu SF, Erickson H, Vandlen R. FRET Reagent Reveals the Intracellular Processing of Peptide-Linked Antibody–Drug Conjugates. Bioconjug Chem 2018; 29:2468-2477. [DOI: 10.1021/acs.bioconjchem.8b00362] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Byoung-Chul Lee
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Cecile Chalouni
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Sophia Doll
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
- Department of Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Sam C. Nalle
- Department of Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Martine Darwish
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Siao Ping Tsai
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Katherine R. Kozak
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Geoffrey Del-Rosario
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shang-Fan Yu
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hans Erickson
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard Vandlen
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
178
|
Buecheler JW, Winzer M, Tonillo J, Weber C, Gieseler H. Impact of Payload Hydrophobicity on the Stability of Antibody–Drug Conjugates. Mol Pharm 2018; 15:2656-2664. [DOI: 10.1021/acs.molpharmaceut.8b00177] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Jakob W. Buecheler
- Division of Pharmaceutics, Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Discovery and Development Technologies, Merck KGaA, 64293 Darmstadt, Germany
| | - Matthias Winzer
- Discovery and Development Technologies, Merck KGaA, 64293 Darmstadt, Germany
| | - Jason Tonillo
- Discovery and Development Technologies, Merck KGaA, 64293 Darmstadt, Germany
| | - Christian Weber
- Discovery and Development Technologies, Merck KGaA, 64293 Darmstadt, Germany
| | | |
Collapse
|
179
|
Consuegra-Fernández M, Lin F, Fox DA, Lozano F. Clinical and experimental evidence for targeting CD6 in immune-based disorders. Autoimmun Rev 2018. [DOI: 10.1016/j.autrev.2017.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
180
|
Harris KE, Aldred SF, Davison LM, Ogana HAN, Boudreau A, Brüggemann M, Osborn M, Ma B, Buelow B, Clarke SC, Dang KH, Iyer S, Jorgensen B, Pham DT, Pratap PP, Rangaswamy US, Schellenberger U, van Schooten WC, Ugamraj HS, Vafa O, Buelow R, Trinklein ND. Sequence-Based Discovery Demonstrates That Fixed Light Chain Human Transgenic Rats Produce a Diverse Repertoire of Antigen-Specific Antibodies. Front Immunol 2018; 9:889. [PMID: 29740455 PMCID: PMC5928204 DOI: 10.3389/fimmu.2018.00889] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/10/2018] [Indexed: 11/13/2022] Open
Abstract
We created a novel transgenic rat that expresses human antibodies comprising a diverse repertoire of heavy chains with a single common rearranged kappa light chain (IgKV3-15-JK1). This fixed light chain animal, called OmniFlic, presents a unique system for human therapeutic antibody discovery and a model to study heavy chain repertoire diversity in the context of a constant light chain. The purpose of this study was to analyze heavy chain variable gene usage, clonotype diversity, and to describe the sequence characteristics of antigen-specific monoclonal antibodies (mAbs) isolated from immunized OmniFlic animals. Using next-generation sequencing antibody repertoire analysis, we measured heavy chain variable gene usage and the diversity of clonotypes present in the lymph node germinal centers of 75 OmniFlic rats immunized with 9 different protein antigens. Furthermore, we expressed 2,560 unique heavy chain sequences sampled from a diverse set of clonotypes as fixed light chain antibody proteins and measured their binding to antigen by ELISA. Finally, we measured patterns and overall levels of somatic hypermutation in the full B-cell repertoire and in the 2,560 mAbs tested for binding. The results demonstrate that OmniFlic animals produce an abundance of antigen-specific antibodies with heavy chain clonotype diversity that is similar to what has been described with unrestricted light chain use in mammals. In addition, we show that sequence-based discovery is a highly effective and efficient way to identify a large number of diverse monoclonal antibodies to a protein target of interest.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Biao Ma
- Teneobio, Inc., Menlo Park, CA, United States
| | | | | | | | | | | | - Duy T Pham
- Teneobio, Inc., Menlo Park, CA, United States
| | | | | | | | | | | | - Omid Vafa
- Teneobio, Inc., Menlo Park, CA, United States
| | | | | |
Collapse
|
181
|
Dantas E, Erra Díaz F, Pereyra Gerber P, Merlotti A, Varese A, Ostrowski M, Sabatté J, Geffner J. Low pH impairs complement-dependent cytotoxicity against IgG-coated target cells. Oncotarget 2018; 7:74203-74216. [PMID: 27716623 PMCID: PMC5342046 DOI: 10.18632/oncotarget.12412] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 09/20/2016] [Indexed: 12/25/2022] Open
Abstract
Local acidosis is a common feature of allergic, vascular, autoimmune, and cancer diseases. However, few studies have addressed the effect of extracellular pH on the immune response. Here, we analyzed whether low pH could modulate complement-dependent cytotoxicity (CDC) against IgG-coated cells. Using human serum as a complement source, we found that extracellular pH values of 5.5 and 6.0 strongly inhibit CDC against either B lymphoblast cell lines coated with the chimeric anti-CD20 mAb rituximab or PBMCs coated with the humanized anti-CD52 mAb alemtuzumab. Suppression of CDC by low pH was observed either in cells suspended in culture medium or in whole blood assays. Interestingly, not only CDC against IgG-coated cells, but also the activation of the complement system induced by the alternative and lectin pathways was prevented by low pH. Tumor-targeting mAbs represent one of the most successful tools for cancer therapy, however, the use of mAb monotherapy has only modest effects on solid tumors. Our present results suggest that severe acidosis, a hallmark of solid tumors, might impair complement-mediated tumor destruction directed by mAb.
Collapse
Affiliation(s)
- Ezequiel Dantas
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Fernando Erra Díaz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Pehuén Pereyra Gerber
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Antonela Merlotti
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Augusto Varese
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Matías Ostrowski
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Juan Sabatté
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), CONICET, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| |
Collapse
|
182
|
Adotevi O, Godet Y, Galaine J, Lakkis Z, Idirene I, Certoux JM, Jary M, Loyon R, Laheurte C, Kim S, Dormoy A, Pouthier F, Barisien C, Fein F, Tiberghien P, Pivot X, Valmary-Degano S, Ferrand C, Morel P, Delabrousse E, Borg C. In situ delivery of allogeneic natural killer cell (NK) combined with Cetuximab in liver metastases of gastrointestinal carcinoma: A phase I clinical trial. Oncoimmunology 2018; 7:e1424673. [PMID: 29721386 DOI: 10.1080/2162402x.2018.1424673] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/16/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023] Open
Abstract
Despite successful introduction of NK-based cellular therapy in the treatment of myeloid leukemia, the potential use of NK alloreactivity in solid malignancies is still elusive. We performed a phase I clinical trial to assess the safety and efficacy of in situ delivery of allogeneic NK cells combined with cetuximab in liver metastasis of gastrointestinal origin. The conditioning chemotherapy was administrated before the allogeneic NK cells injection via hepatic artery. Three escalating doses were tested (3.106, 8.106 and 12.106 NK cells/kg) following by a high-dose interleukin-2 (IL-2). Cetuximab was administered intravenously every week for 7 weeks. Nine patients with liver metastases of colorectal or pancreatic cancers were included, three per dose level. Hepatic artery injection was successfully performed in all patients with no report of dose-limiting toxicity. Two patients had febrile aplasia requiring a short-term antibiotherapy. Grade 3/4 anemia and thrombopenia were also observed related to the chemotherapy. Objective clinical responses were documented in 3 patients and among them 2 occurred in patients injected with cell products harboring two KIR ligand mismatches and one in a patient with one KIR ligand mismatch. Immune monitoring revealed that most patients presented an increase but transient of IL-15 and IL-7 cytokines levels one week after chemotherapy. Furthermore, a high expansion of FoxP3+regulatory T cells and PD-1+ T cells was observed in all patients, related to IL-2 administration. Our results demonstrated that combining allogeneic NK cells transfer via intra-hepatic artery, cetuximab and a high-dose IL-2 is feasible, well tolerated and may result in clinical responses.
Collapse
Affiliation(s)
- O Adotevi
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - Y Godet
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - J Galaine
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - Z Lakkis
- University Hospital of Besançon, Department of Gastrointestinal and liver surgery, F-25000 Besançon, France
| | - I Idirene
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - J M Certoux
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - M Jary
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - R Loyon
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - C Laheurte
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,Etablissement Français du Sang Bourgogne Franche-Comté, plateforme de BioMonitoring, F-25000 Besançon, France.,INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center in Biotherapy, F-25000, Besançon, France
| | - S Kim
- University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - A Dormoy
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - F Pouthier
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - C Barisien
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - F Fein
- University Hospital of Besançon, Department of Gastroenterology, F-25000 Besançon, France
| | - P Tiberghien
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - X Pivot
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| | - S Valmary-Degano
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Pathology, F-25000 Besançon, France
| | - C Ferrand
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France
| | - P Morel
- Etablissement Français du Sang Bourgogne Franche-Comté, F-25000 Besançon, France
| | - E Delabrousse
- University Hospital of Besançon, Department of Radiology, F-25000, Besançon, France
| | - C Borg
- University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, F-25000 Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, F-25000 Besançon, France
| |
Collapse
|
183
|
Conibear AC, Pötgens AJG, Thewes K, Altdorf C, Hilzendeger C, Becker CFW. Synthetic Cancer-Targeting Innate Immune Stimulators Give Insights into Avidity Effects. Chembiochem 2018; 19:459-469. [PMID: 29230922 DOI: 10.1002/cbic.201700522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Indexed: 11/08/2022]
Abstract
Multispecific and multivalent antibodies are seen as promising cancer therapeutics, and numerous antibody fragments and derivatives have been developed to exploit avidity effects that result in increased selectivity. Most of these multispecific and multivalent antibody strategies make use of recombinant expression of antigen-binding modules. In contrast, chemical synthesis and chemoselective ligations can be used to generate a variety of molecules with different numbers and combinations of binding moieties in a modular and homogeneous fashion. In this study we synthesized a series of targeted immune system engagers (ISErs) by using solid-phase peptide synthesis and chemoselective ligations. To explore avidity effects, we constructed molecules bearing different numbers and combinations of two "binder" peptides that target ephrin A2 and integrin α3 receptors and an "effector" peptide that binds to formyl peptide receptors and stimulates an immune response. We investigated various strategies for generating multivalent and multispecific targeted innate immune stimulators and studied their activities in terms of binding to cancer cells and stimulation of immune cells. This study gives insights into the influence that multivalency and receptor density have on avidity effects and is useful for the design of potential anticancer therapeutics.
Collapse
Affiliation(s)
- Anne C Conibear
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Strasse 38, 1090, Vienna, Austria
| | - André J G Pötgens
- Syntab Therapeutics GmbH, ZBMT, Pauwelstrasse 17, 52074, Aachen, Germany
| | - Karine Thewes
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Strasse 38, 1090, Vienna, Austria
| | - Claudia Altdorf
- Syntab Therapeutics GmbH, ZBMT, Pauwelstrasse 17, 52074, Aachen, Germany
| | | | - Christian F W Becker
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Strasse 38, 1090, Vienna, Austria
| |
Collapse
|
184
|
Jacob L, Sawma P, Garnier N, Meyer LAT, Fritz J, Hussenet T, Spenlé C, Goetz J, Vermot J, Fernandez A, Baumlin N, Aci-Sèche S, Orend G, Roussel G, Crémel G, Genest M, Hubert P, Bagnard D. Inhibition of PlexA1-mediated brain tumor growth and tumor-associated angiogenesis using a transmembrane domain targeting peptide. Oncotarget 2018; 7:57851-57865. [PMID: 27506939 PMCID: PMC5295395 DOI: 10.18632/oncotarget.11072] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 07/21/2016] [Indexed: 11/25/2022] Open
Abstract
The neuropilin-plexin receptor complex regulates tumor cell migration and proliferation and thus is an interesting therapeutic target. High expression of neuropilin-1 is indeed associated with a bad prognosis in glioma patients. Q-RTPCR and tissue-array analyses showed here that Plexin-A1 is highly expressed in glioblastoma and that the highest level of expression correlates with the worse survival of patients. We next identified a developmental and tumor-associated pro-angiogenic role of Plexin-A1. Hence, by using molecular simulations and a two-hybrid like assay in parallel with biochemical and cellular assays we developed a specific Plexin-A1 peptidic antagonist disrupting transmembrane domain-mediated oligomerization of the receptor and subsequent signaling and functional activity. We found that this peptide exhibits anti-tumor activity in vivo on different human glioblastoma models including glioma cancer stem cells. Thus, screening Plexin-A1 expression and targeting Plexin-A1 in glioblastoma patients exhibit diagnostic and therapeutic value.
Collapse
Affiliation(s)
- Laurent Jacob
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Paul Sawma
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires (LISM), UMR 7255, CNRS-Aix Marseille Université, Marseille, France
| | - Norbert Garnier
- Centre de Biophysique Moléculaire, UPR 4301, CNRS, Affiliated to the University of Orléans, Orléans, France
| | - Lionel A T Meyer
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Justine Fritz
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Thomas Hussenet
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Caroline Spenlé
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jacky Goetz
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.,Institute of Genetics and Molecular and Cellular Biology (IGBMC), CNRS/INSERM/UDS, Illkirch, France
| | - Julien Vermot
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), CNRS/INSERM/UDS, Illkirch, France
| | - Aurore Fernandez
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Nadège Baumlin
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Samia Aci-Sèche
- Centre de Biophysique Moléculaire, UPR 4301, CNRS, Affiliated to the University of Orléans, Orléans, France.,Current address: Institut de Chimie Organique et Analytique UMR, Université d'Orléans, Orléans, France
| | - Gertraud Orend
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Guy Roussel
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Gérard Crémel
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Monique Genest
- Centre de Biophysique Moléculaire, UPR 4301, CNRS, Affiliated to the University of Orléans, Orléans, France
| | - Pierre Hubert
- Laboratoire d’Ingénierie des Systèmes Macromoléculaires (LISM), UMR 7255, CNRS-Aix Marseille Université, Marseille, France
| | - Dominique Bagnard
- MN3T Team, INSERM U1109, Strasbourg, France.,Université de Strasbourg, Strasbourg, France.,LabEx Medalis, Université de Strasbourg, Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
185
|
Identification of an HSP90 modulated multi-step process for ERBB2 degradation in breast cancer cells. Oncotarget 2018; 7:85411-85429. [PMID: 27863425 PMCID: PMC5356745 DOI: 10.18632/oncotarget.13392] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/28/2016] [Indexed: 12/24/2022] Open
Abstract
The receptor tyrosine kinase ERBB2 interacts with HSP90 and is overexpressed in aggressive breast cancers. Therapeutic HSP90 inhibitors, i.e. Geldanamycin (GA), target ERBB2 to degradation. We have previously shown that HSP90 is responsible for the missorting of recycling ERBB2 to degradation compartments. In this study, we used biochemical, immunofluorescence and electron microscopy techniques to demonstrate that in SKBR3 human breast cancer cells, GA strongly induces polyubiquitination and internalization of the full-length p185-ERBB2, and promotes its cleavage, with the formation of a p116-ERBB2 form in EEA1-positive endosomes (EE). p116-ERBB2 corresponds to a non-ubiquitinated, signaling-impaired, membrane-bound fragment, which is readily sorted to lysosomes and degraded. To define the sequence of events leading to p116-ERBB2 degradation, we first blocked the EE maturation/trafficking to late endosomes/lysosomes with wortmannin, and found an increase in GA-dependent formation of p116-ERBB2; we then inhibited the proteasome activity with MG-132 or lactacystin, and observed an efficient block of p185-ERBB2 cleavage, and its accumulation in EE, suggesting that p185-ERBB2 polyubiquitination is necessary for proteasome-dependent p116-ERBB2 generation occurring in EE. As polyubiquitination has also been implicated in autophagy-mediated degradation of ERBB2 under different experimental conditions, we exploited this possibility and demonstrate that GA strongly inhibits early autophagy, and reduces the levels of the autophagy markers atg5-12 and LC3-II, irrespective of GA-induced ERBB2 polyubiquitination, ruling out a GA-dependent autophagic degradation of ERBB2. In conclusion, we propose that HSP90 inhibition fosters ERBB2 polyubiquitination and proteasome-dependent generation of a non-ubiquitinated and inactive p116-ERBB2 form in EE, which is trafficked from altered EE to lysosomes.
Collapse
|
186
|
Chalouni C, Doll S. Fate of Antibody-Drug Conjugates in Cancer Cells. J Exp Clin Cancer Res 2018; 37:20. [PMID: 29409507 PMCID: PMC5802061 DOI: 10.1186/s13046-017-0667-1] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 12/15/2017] [Indexed: 02/08/2023] Open
Abstract
Antibody-Drug Conjugates (ADCs) are a class of cancer therapeutics that combines antigen specificity and potent cytotoxicity in a single molecule as they are comprised of an engineered antibody linked chemically to a cytotoxic drug. Four ADCs have received approval by the Food and Drug Administration (FDA) and the European Medicine Agency (EMA) and can be prescribed for metastatic conditions while around 60 ADCs are currently enrolled in clinical trials. The efficacy of an ADC greatly relies on its intracellular trafficking and processing of its components to trigger tumor cell death. A limited number of studies have addressed these critical processes that both challenge and help foster the design of ADCs. This review highlights those mechanisms and their relevance for future development of ADCs as cancer therapeutics.
Collapse
|
187
|
Microfluidic single-cell technology in immunology and antibody screening. Mol Aspects Med 2018; 59:47-61. [DOI: 10.1016/j.mam.2017.09.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 11/20/2022]
|
188
|
Kim TK, Park CS, Jang J, Kim MR, Na HJ, Lee K, Kim HJ, Heo K, Yoo BC, Kim YM, Lee JW, Kim SJ, Kim ES, Kim DY, Cha K, Lee TG, Lee S. Inhibition of VEGF-dependent angiogenesis and tumor angiogenesis by an optimized antibody targeting CLEC14a. Mol Oncol 2018; 12:356-372. [PMID: 29316206 PMCID: PMC5830631 DOI: 10.1002/1878-0261.12169] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/12/2022] Open
Abstract
The C‐type lectin‐like domain of CLEC14a (CLEC14a‐C‐type lectin‐like domain [CTLD]) is a key domain that mediates endothelial cell–cell contacts in angiogenesis. However, the role of CLEC14a‐CTLD in pathological angiogenesis has not yet been clearly elucidated. In this study, through complementarity‐determining region grafting, consecutive deglycosylation, and functional isolation, we generated a novel anti‐angiogenic human monoclonal antibody that specifically targets CLEC14a‐CTLD and that shows improved stability and homogeneity relative to the parental antibody. We found that this antibody directly inhibits CLEC14a‐CTLD‐mediated endothelial cell–cell contact and simultaneously downregulates expression of CLEC14a on the surface of endothelial cells. Using various in vitro and in vivo functional assays, we demonstrated that this antibody effectively suppresses vascular endothelial growth factor (VEGF)‐dependent angiogenesis and tumor angiogenesis of SNU182 human hepatocellular carcinoma, CFPAC‐1 human pancreatic cancer, and U87 human glioma cells. Furthermore, we also found that this antibody significantly inhibits tumor angiogenesis of HCT116 and bevacizumab‐adapted HCT116 human colorectal cancer cells. These findings suggest that antibody targeting of CLEC14a‐CTLD has the potential to suppress VEGF‐dependent angiogenesis and tumor angiogenesis and that CLEC14a‐CTLD may be a novel anti‐angiogenic target for VEGF‐dependent angiogenesis and tumor angiogenesis.
Collapse
Affiliation(s)
- Taek-Keun Kim
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Chang Sik Park
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Jihye Jang
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Mi Ra Kim
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Hee-Jun Na
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Kangseung Lee
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| | - Hyun Jung Kim
- Research Institute, National Cancer Center, Goyang, South Korea
| | - Kyun Heo
- Research Institute, National Cancer Center, Goyang, South Korea
| | - Byong Chul Yoo
- Research Institute, National Cancer Center, Goyang, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, Kangwon National University, Chuncheon, South Korea
| | - Je-Wook Lee
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Su Jin Kim
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Eun Sung Kim
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Dae Young Kim
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Kiweon Cha
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Tae Gyu Lee
- New Drug Development Center, Osong Medical Innovation Foundation, South Korea
| | - Sukmook Lee
- Scripps Korea Antibody Institute, Chuncheon, South Korea
| |
Collapse
|
189
|
Chen X, Nagai Y, Zhu Z, Ruan H, Peehl DM, Greene MI, Zhang H. A spliced form of CD44 expresses the unique glycan that is recognized by the prostate cancer specific antibody F77. Oncotarget 2018; 9:3631-3640. [PMID: 29423071 PMCID: PMC5790488 DOI: 10.18632/oncotarget.23341] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/26/2017] [Indexed: 01/08/2023] Open
Abstract
Prostate cancer is the most common cancer occurring in men in the United States. The monoclonal antibody F77 that was originally developed in our laboratory recognizes mainly glycolipids as well as O-linked glycosylation on proteins in prostate cancer cells. We have identified a spliced form of glycoprotein CD44 as one critical protein expressing the F77 antigen. The F77-specific glycosylation occurs on multiple potential glycosylation sites on the CD44 protein encoded by the fourteenth exon. CD44 is a tumor stem cell marker and is known to induce tumor stemness and metastasis. Knockdown of CD44 or FUT1 genes dramatically reduced F77-induced apoptosis in prostate cancer cell lines. We developed an ELISA using both a CD44 antibody and F77 to identify the special form of glycosylated CD44 from prostate cancer cells as well as from serum samples of prostate cancer patients. These results reveal a CD44-dependent mechanism for F77 to induce tumor cell apoptosis, and a new strategy for the detection of glycosylated CD44 proteins secreted by prostate cancer cells.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yasuhiro Nagai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiqiang Zhu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hang Ruan
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donna M. Peehl
- Department of Urology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark I. Greene
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongtao Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
190
|
LC–MS Challenges in Characterizing and Quantifying Monoclonal Antibodies (mAb) and Antibody-Drug Conjugates (ADC) in Biological Samples. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s40495-017-0118-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
191
|
Wan J, Sun L, Wu P, Wang F, Guo J, Cheng J, Wang C. Synthesis of indocyanine green functionalized comblike poly(aspartic acid) derivatives for enhanced cancer cell ablation by targeting the endoplasmic reticulum. Polym Chem 2018. [DOI: 10.1039/c7py01994g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PTX-loaded comblike polymer PAsp-g-(PEG-ICG) micelles can effectively kill cancer cells via elevated endoplasmic reticulum stress under laser irradiation.
Collapse
Affiliation(s)
- Jiaxun Wan
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P.R. China
| | - Luyan Sun
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P.R. China
| | - Pan Wu
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P.R. China
| | - Fang Wang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P.R. China
| | - Jia Guo
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P.R. China
| | - Jianjun Cheng
- Department of Materials Science and Engineering
- University of Illinois at Urbana–Champaign
- Urbana
- USA
| | - Changchun Wang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- P.R. China
| |
Collapse
|
192
|
YAMADA T, MIZUNO H, Zhe MIN J, TOYO’OKA T, TODOROKI K. High Sensitivity and Precision High-Temperature Reversed-Phase LC Analysis of Bevacizumab for Intact Bioanalysis of Therapeutic Monoclonal Antibodies. CHROMATOGRAPHY 2018. [DOI: 10.15583/jpchrom.2017.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
| | - Hajime MIZUNO
- School of Pharmaceutical Sciences, University of Shizuoka
| | - Jun Zhe MIN
- Key Laboratory for Natural Resource of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University
| | | | | |
Collapse
|
193
|
Elamin KM, Yamashita Y, Higashi T, Motoyama K, Arima H. Supramolecular Complex of Methyl-β-cyclodextrin with Adamantane-Grafted Hyaluronic Acid as a Novel Antitumor Agent. Chem Pharm Bull (Tokyo) 2017; 66:277-285. [PMID: 29269686 DOI: 10.1248/cpb.c17-00824] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Methyl-β-cyclodextrin (M-β-CyD) exhibits cytotoxic activity, and has the potentials as an antitumor agent. However, a tumor-selectivity of M-β-CyD is low, leading to low antitumor activity and the adverse effects. Meanwhile, hyaluronic acid (HA) is known as a promising tumor targeting ligand, because various cancer cells overexpress CD44, a HA-binding glycoprotein. In the present study, to develop a tumor-selective delivery system for M-β-CyD, we designed a supramolecular complex of M-β-CyD with adamantane-grafted HA (Ad-HA/M-β-CyD) and evaluated it as a tumor-selective antitumor agent. M-β-CyD formed a stable complex with Ad-HA (Kc>104 M-1). In addition, Ad-HA/M-β-CyD formed slightly a negative-charged nanoparticle with ca. 140 nm of a particle size, indicating the favorable physicochemical properties for antitumor agents. Ad-HA/M-β-CyD showed the superior cytotoxic activity via CD44-mediated endosomal pathways in HCT116 cells (CD44(+)), a human colon cancer cell line. In addition, cytotoxic activity of Ad-HA/M-β-CyD was induced by apoptosis. These results suggest that Ad-HA/M-β-CyD has the potentials as a tumor-selective supramolecular antitumor agent.
Collapse
Affiliation(s)
- Khaled Mohamed Elamin
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Yamashita
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Taishi Higashi
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Keiichi Motoyama
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Hidetoshi Arima
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University.,Program for Leading Graduate Schools "HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program," Kumamoto University
| |
Collapse
|
194
|
Qi R, Wang Y, Bruno PM, Xiao H, Yu Y, Li T, Lauffer S, Wei W, Chen Q, Kang X, Song H, Yang X, Huang X, Detappe A, Matulonis U, Pepin D, Hemann MT, Birrer MJ, Ghoroghchian PP. Nanoparticle conjugates of a highly potent toxin enhance safety and circumvent platinum resistance in ovarian cancer. Nat Commun 2017; 8:2166. [PMID: 29255160 PMCID: PMC5735131 DOI: 10.1038/s41467-017-02390-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/21/2017] [Indexed: 12/19/2022] Open
Abstract
Advanced-stage epithelial ovarian cancers are amongst the most difficult to treat tumors and have proven to be refractory to most cytotoxic, molecularly targeted, or immunotherapeutic approaches. Here, we report that nanoparticle-drug conjugates (NDCs) of monomethyl auristatin E (MMAE) significantly increase loading on a per-vehicle basis as compared to antibody-drug conjugates (ADCs). Their intraperitoneal administration enabled triggered release of the active MMAE toxin to inhibit tumor growth and to extend animal survival to >90 days in a cell-line xenograft model of disseminated ovarian cancer. In a patient-derived xenograft model of advanced-stage and platinum-resistant ovarian cancer, an MMAE-based NDC doubled the duration of tumor growth inhibition as compared to cisplatin. NDCs of highly potent toxins thus introduce a translatable platform that may be exploited to maximize the safety and efficacy of cytotoxic chemotherapies, combining the best features of ADCs with those of nanoparticle-based therapeutics. Improving the safety and efficacy of chemotherapeutics will help to enhance their effects. Here, the authors show that intraperitoneal delivery of nanoparticle conjugates of a potent toxin prolongs tumor inhibition and survival as compared to cisplatin in advanced-stage and platinum-resistant ovarian cancer mouse models.
Collapse
Affiliation(s)
- Ruogu Qi
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Yongheng Wang
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Peter M Bruno
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Haihua Xiao
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Yingjie Yu
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Ting Li
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA.,Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Sam Lauffer
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Wei Wei
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Qixian Chen
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Xiang Kang
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Haiqin Song
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Xi Yang
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Xing Huang
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Alexandre Detappe
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA.,Dana Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.,Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
| | - Ursula Matulonis
- Dana Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.,Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
| | - David Pepin
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.,Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA
| | - Michael T Hemann
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA
| | - Michael J Birrer
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA. .,Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA.
| | - P Peter Ghoroghchian
- Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA, 02139, USA. .,Dana Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA. .,Harvard Medical School, 25 Shattuck Street, Boston, MA, 02115, USA.
| |
Collapse
|
195
|
High-throughput sequencing of the immune repertoire in oncology: Applications for clinical diagnosis, monitoring, and immunotherapies. Cancer Lett 2017; 416:42-56. [PMID: 29247824 DOI: 10.1016/j.canlet.2017.12.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/11/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022]
Abstract
The diagnostic, monitoring and therapeutic options for cancers currently remain limited. These limitations represent a large threat to human health. Adaptive immunity, which is dependent on diverse repertoires of B cell receptors (BCRs) and T cell receptors (TCRs), plays a critical role in the anti-tumor immune response. Modulation and surveillance of adaptive immunity has become a powerful weapon to combat cancers. Recently, the high-throughput sequencing of immune repertoire (HTS-IR) technology, which provides a robust tool for deep sequencing repertoires of BCRs or TCRs, has been applied in the development of tumor biomarkers and immunotherapeutics for cancers. This review will first provide an overview of the advancement of HTS-IR technology at the population-cell and single-cell levels. It will then provide a current summary of the applications of HTS-IR technology in the diagnosis and monitoring of minimal residual disease (MRD), focusing on immune reconstitution after the treatment of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in B/T-cell malignancies, and the precise detection of tumor-infiltrating lymphocytes (TILs) in non-B/T-cell malignancies. Finally, current advances of HTS-IR technology in cancer immunotherapeutic applications, such as therapeutic antibodies, CAR-T cell based-adoptive immunotherapies, and neoantigen-specific TCR-T cell-based adoptive immunotherapies, will be introduced.
Collapse
|
196
|
Zhang X, Sun L. Anaphylatoxin C3a: A potential biomarker for esophageal cancer diagnosis. Mol Clin Oncol 2017; 8:315-319. [PMID: 29435296 DOI: 10.3892/mco.2017.1524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/28/2017] [Indexed: 12/30/2022] Open
Abstract
Esophageal carcinoma is a common malignancy worldwide, with a low 5-year survival rate. As the majority of cases are diagnosed at an advanced stage, there is an urgent need for an effective biomarker for early diagnosis of esophageal cancer patients. Surface-enhanced laser desorption ionization time-of-flight mass spectrometry (SELDI-TOF-MS) was applied to detect the serum protein expression in esophageal cancer patients using ProteinChip software, and the results were analyzed and screened using Biomarker Patterns and SPSS16.0 software. The ELISA method was conducted to determine the concentration of anaphylatoxin C3a, which is one of the complement proteins, in the serum of esophageal cancer patients and non-esophageal cancer participants. A total of 144 effective differential expression protein peaks in the window of 1-10 kDa were obtained (P<0.05). M/Z 8,926.478 (P<10-6) protein peak was employed as the diagnostic biomarker for esophageal carcinoma. This established diagnostic biomarker has a sensitivity of 95% (19/20) and an accuracy of 100% (19/19) for positive prediction. The results suggested that anaphylatoxin C3a may be a promising biomarker in the diagnosis of esophageal carcinoma, and may play a key role in promoting esophageal carcinogenesis.
Collapse
Affiliation(s)
- Xu Zhang
- School of Pharmacy, Yancheng Teachers University, Yancheng, Jiangsu 224002, P.R. China
| | - Lingzhi Sun
- School of Pharmacy, Yancheng Teachers University, Yancheng, Jiangsu 224002, P.R. China
| |
Collapse
|
197
|
Alam ME, Geng SB, Bender C, Ludwig SD, Linden L, Hoet R, Tessier PM. Biophysical and Sequence-Based Methods for Identifying Monovalent and Bivalent Antibodies with High Colloidal Stability. Mol Pharm 2017; 15:150-163. [PMID: 29154550 DOI: 10.1021/acs.molpharmaceut.7b00779] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In vitro antibody discovery and/or affinity maturation are often performed using antibody fragments (Fabs), but most monovalent Fabs are reformatted as bivalent IgGs (monoclonal antibodies, mAbs) for therapeutic applications. One problem related to reformatting antibodies is that the bivalency of mAbs can lead to increased antibody self-association and poor biophysical properties (e.g., reduced antibody solubility and increased viscosity). Therefore, it is important to identify monovalent Fabs early in the discovery and/or optimization process that will display favorable biophysical properties when reformatted as bivalent mAbs. Here we demonstrate a facile approach for evaluating Fab self-association in a multivalent assay format that is capable of identifying antibodies with low self-association and favorable colloidal properties when reformatted as bivalent mAbs. Our approach (self-interaction nanoparticle spectroscopy, SINS) involves immobilizing Fabs on gold nanoparticles in a multivalent format (multiple Fabs per nanoparticle) and evaluating their self-association behavior via shifts in the plasmon wavelength or changes in the absorbance values. Importantly, we find that SINS measurements of Fab self-association are correlated with self-interaction measurements of bivalent mAbs and are useful for identifying antibodies with favorable biophysical properties. Moreover, the significant differences in the levels of self-association detected for Fabs and mAbs with similar frameworks can be largely explained by the physicochemical properties of the complementarity-determining regions (CDRs). Comparison of the properties of the CDRs in this study relative to those of approved therapeutic antibodies reveals several key factors (net charge, fraction of charged residues, and presence of self-interaction motifs) that strongly influence antibody self-association behavior. Increased positive charge in the CDRs was observed to correlate with increased risk of high self-association for the mAbs in this study and clinical-stage antibodies. We expect that these findings will be useful for improving the development of therapeutic antibodies that are well suited for high concentration applications.
Collapse
Affiliation(s)
- Magfur E Alam
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - Steven B Geng
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - Christian Bender
- Pharmaceuticals, Bayer AG , Nattermannallee 1, Cologne 50829, Germany
| | - Seth D Ludwig
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York 12180, United States
| | - Lars Linden
- Pharmaceuticals, Bayer AG , Aprather Weg 18A, Wuppertal 42117, Germany
| | - Rene Hoet
- Pharmaceuticals, Bayer AG , Nattermannallee 1, Cologne 50829, Germany
| | - Peter M Tessier
- Isermann Department of Chemical & Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute , Troy, New York 12180, United States.,Departments of Chemical Engineering, Pharmaceutical Sciences and Biomedical Engineering, Biointerfaces Institute, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
198
|
Tremble LF, Forde PF, Soden DM. Clinical evaluation of macrophages in cancer: role in treatment, modulation and challenges. Cancer Immunol Immunother 2017; 66:1509-1527. [PMID: 28948324 PMCID: PMC11028704 DOI: 10.1007/s00262-017-2065-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/13/2017] [Indexed: 12/22/2022]
Abstract
The focus of immunotherapeutics has been placed firmly on anti-tumour T cell responses. Significant progress has been made in the treatment of both local and systemic malignancies, but low response rates and rising toxicities are limiting this approach. Advancements in the understanding of tumour immunology are opening up a new range of therapeutic targets, including immunosuppressive factors in the tumour microenvironment. Macrophages are a heterogeneous group of cells that have roles in innate and adaptive immunity and tissue repair, but become co-opted by tumours to support tumour growth, survival, metastasis and immunosuppression. Macrophages also support tumour resistance to conventional therapy. In preclinical models, interference with macrophage migration, macrophage depletion and macrophage re-education have all been shown to reduce tumour growth and support anti-tumour immune responses. Here we discuss the role of macrophages in prognosis and sensitivity to therapy, while examining the significant progress which has been made in modulating the behaviour of these cells in cancer patients.
Collapse
Affiliation(s)
- Liam Friel Tremble
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland.
| | - Patrick F Forde
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland
| | - Declan M Soden
- Cork Cancer Research Centre, Western Gateway Building, University College Cork, Western Road, Cork, Ireland
| |
Collapse
|
199
|
Abstract
Over the past two decades, hundreds of new somatic mutations have been identified in tumours, and a few dozen novel cancer therapeutics that selectively target these mutated oncoproteins have entered clinical practice. This development has resulted in clinical breakthroughs for a few tumour types, but more commonly patients' overall survival has not improved because of the development of drug resistance. Furthermore, only a very limited number of oncoproteins, largely protein kinases, are successfully targeted, whereas most non-kinase oncoproteins inside cancer cells remain untargeted. Engineered small protein inhibitors offer great promise in targeting a larger variety of oncoproteins with better efficacy and higher selectivity. In this article, I focus on a promising class of synthetic binding proteins, termed monobodies, that we have shown to inhibit previously untargetable protein-protein interactions in different oncoproteins. I will discuss the great promise alongside the technical challenges inherent in converting monobodies from potent pre-clinical target validation tools to next-generation protein-based therapeutics.
Collapse
Affiliation(s)
- Oliver Hantschel
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
200
|
Lee J, Shin YJ, Lee K, Cho HJ, Sa JK, Lee SY, Kim SH, Lee J, Yoon Y, Nam DH. Anti-SEMA3A Antibody: A Novel Therapeutic Agent to Suppress Glioblastoma Tumor Growth. Cancer Res Treat 2017; 50:1009-1022. [PMID: 29129044 PMCID: PMC6056981 DOI: 10.4143/crt.2017.315] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/30/2017] [Indexed: 11/21/2022] Open
Abstract
Purpose Glioblastoma (GBM) is classified as one of the most aggressive and lethal brain tumor. Great strides have been made in understanding the genomic and molecular underpinnings of GBM, which translated into development of new therapeutic approaches to combat such deadly disease. However, there are only few therapeutic agents that can effectively inhibit GBM invasion in a clinical framework. In an effort to address such challenges, we have generated anti-SEMA3A monoclonal antibody as a potential therapeutic antibody against GBM progression. Materials and Methods We employed public glioma datasets, Repository of Molecular Brain Neoplasia Data and The Cancer Genome Atlas, to analyze SEMA3AmRNA expression in human GBM specimens. We also evaluated for protein expression level of SEMA3A via tissue microarray (TMA) analysis. Cell migration and proliferation kinetics were assessed in various GBM patient-derived cells (PDCs) and U87-MG cell-line for SEMA3A antibody efficacy. GBM patient-derived xenograft (PDX) models were generated to evaluate tumor inhibitory effect of anti-SEMA3A antibody in vivo. Results By combining bioinformatics and TMA analysis, we discovered that SEMA3A is highly expressed in human GBM specimens compared to non-neoplastic tissues. We developed three different anti-SEMA3A antibodies, in fully human IgG form, through screening phage-displayed synthetic antibody library using a classical panning method. Neutralization of SEMA3A significantly reduced migration and proliferation capabilities of PDCs and U87-MG cell line in vitro. In PDX models, treatment with anti-SEMA3A antibody exhibited notable tumor inhibitory effect through down-regulation of cellular proliferative kinetics and tumor-associated macrophages recruitment. Conclusion In present study, we demonstrated tumor inhibitory effect of SEMA3A antibody in GBM progression and present its potential relevance as a therapeutic agent in a clinical framework.
Collapse
Affiliation(s)
- Jaehyun Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Yong Jae Shin
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoungmin Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Hee Jin Cho
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jason K Sa
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang-Yun Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Seok-Hyung Kim
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea.,Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeongwu Lee
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yeup Yoon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| | - Do-Hyun Nam
- Institute for Refractory Cancer Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|