An Overview on Vadimezan (DMXAA): the Vascular Disrupting Agent

Total Page:16

File Type:pdf, Size:1020Kb

An Overview on Vadimezan (DMXAA): the Vascular Disrupting Agent See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/322146064 An overview on Vadimezan (DMXAA): The vascular disrupting agent Article in Chemical Biology & Drug Design · December 2017 DOI: 10.1111/cbdd.13166 CITATIONS READS 4 66 5 authors, including: Amir Daei Rana Jahanban-Esfahlan Tabriz University of Medical Sciences Tabriz University of Medical Sciences 6 PUBLICATIONS 10 CITATIONS 42 PUBLICATIONS 456 CITATIONS SEE PROFILE SEE PROFILE Khaled Seidi N Zarghami Tabriz University of Medical Sciences Tabriz University of Medical Sciences 21 PUBLICATIONS 123 CITATIONS 270 PUBLICATIONS 3,731 CITATIONS SEE PROFILE SEE PROFILE Some of the authors of this publication are also working on these related projects: The comparison between the effects of pure curcumin and curcumin-loaded PLGA-PEG nanoparticle on telomerase and TRF1 genes expression in lung cancer cell line View project 3-D printing system to produce my model skin tissue View project All content following this page was uploaded by Amir Daei on 30 January 2018. The user has requested enhancement of the downloaded file. Received: 5 August 2017 | Revised: 29 November 2017 | Accepted: 17 December 2017 DOI: 10.1111/cbdd.13166 REVIEW An overview on Vadimezan (DMXAA): The vascular disrupting agent Amir Daei Farshchi Adli1 | Rana Jahanban-Esfahlan1,2 | Khaled Seidi1 | Sonia Samandari-Rad3,4 | Nosratollah Zarghami1,5,6 1Department of Medical Biotechnology, Faculty of Advanced Vascular disrupting agents (VDAs), a group of cancer remedies, can cause a specific Medical Sciences, Tabriz University of and irreversible destruction of established tumor vessels, and the complete halt of Medical Sciences, Tabriz, Iran blood flow in the tumor. DMXAA (ASA404) or Vadimezan, a flavone- acetic acid- 2 Student Research Committee, Tabriz based drug, is the most promising VDAs that induces a rapid shutdown of blood flow University of Medical Sciences, Tabriz, Iran in tumors but not in normal tissue. The exact mechanism of vascular disruption is 3Faculty of Medicine, Physiology Research Center, Tehran University of Medical unknown; however, proposed direct and indirect mechanisms of action for DMXAA Sciences, Tehran, Iran comprises (i) inducing apoptosis in endothelial cells; (ii) hemorrhagic necrosis and 4 Department of Physiology, Faculty of ischemia in tumor; (iii) release of serotonin (5-HT); (vi) stimulation of innate im- Medicine, Tehran University of Medical mune system; (v) production of inflammatory cytokines, for example TNF, IL- 6, Sciences, Tehran, Iran 5Department of Clinical Biochemistry GCSF, KC, IP- 10, and MCP- 1; (vi) activation of NFκB and p38 (MAPK); (vii) pro- and Laboratory Medicine, Faculty of duction of nitric oxide; and (viii) reducing tumor energetics and membrane turnover. Medicine, Tabriz University of Medical Despite the remarkable results from preclinical and phase I/II, DMXAA has failed in Sciences, Tabriz, Iran phase III clinical trials. The reason for this surprising discrepancy, among others, 6Iranian National Science Foundation, Tehran, Iran was discovered to be STING receptor variations between mice and humans. In this review, the development, the mechanisms of DMXAA action, the clinical trials, the Correspondence combination therapy, and the future of this drug will be discussed. Nosratollah Zarghami, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. KEYWORDS Email: [email protected] ASA404, cancer combination therapy, DMXAA, Vadimezan, vascular disrupting agents (VDAs) [7] 1 | INTRODUCTION antivascular agents. Small antivascular agents also include tubulin binding agents—for example, combretastatin- A4 phos- Cancer is a public health issue and one of the chief causes of phate,[8] OXi4503,[9] and flavonoid derivatives.[10] DMXAA death in the world with about 8.2 million deaths in 2012.[1,2] (5,6- dimethylxanthenone- 4- acetic acid, Vadimezan) is a flavo- The rate of cancer is expected to increase. So finding a new noid derivative that is considered one of the most promising effective remedy is a necessity. In recent years, targeting of antivascular agents.[11] Currently, DMXAA has undergone tumor vessels has been one of the main strategies for cancer several clinical trials. In this review, development, and mech- treatment, due to its unmatched characteristics.[3,4] Solid tu- anism of DMXAA action, the clinical trials, the combination mors require new blood vessels to deliver nutrients and oxy- therapy, and the forthcoming of this drug will be discussed. gen to the cancer cells to grow and survive.[5] As angiogenesis inhibitors interfere with the formation of new blood vessels in solid tumors, antivascular agents target the established vessels 1.1 | DMXAA development in the tumor tissue exclusively.[6] There are two main catego- The production of DMXAA drug was based on the discovery ries of antivascular agents: ligand-dependent agents and small that flavone- 8- acetic acid (FAA) can cause tumor necrosis Chem Biol Drug Des. 2018;1–12. wileyonlinelibrary.com/journal/cbdd © 2017 John Wiley & Sons A/S | 1 2 | DAEI FARSHCHI ADLI ET AL. (Figure 1). The FAA, a flavonoid group—which was initially (DMXAA direct mechanism of action) includes specific synthesized as a non- steroidal anti- inflammatory drug—sur- and irreversible destruction of established tumor vascula- prisingly, demonstrated antitumor properties, as well.[12–14] tures and the complete blockade of tumor blood flow.[14,20] However, the FAA was unsuccessful in clinical trials.[15] This effect is due to the induction of apoptosis in tumor Therefore, the researchers at the Auckland Cancer Society endothelial cells, which, in turn, causes hemorrhagic ne- Research Center (ACSRC) began testing for a more active crosis and ischemia in tumor tissues.[20–22] The destruction form of the FAA and were able to synthesize DMXAA, a de- of tumor vessels can be considered as the first impact of rivative of xanthone and FAA analog. DMXAA has 10 to 15 DMXAA that appears about 30 min after injection.[23,24] times more activity than FAA. The other names of this drug The destruction of tumor vessel through aggregation of are ASA404 and Vadimezan.[16] platelets stimulates and increases the release of serotonin (5- HT) in the plasma.[23,24] Thus, we can consider the sta- ble form of serum serotonin (5HIAA) as an antivascular 1.2 | Proposed mechanism of marker.[25] The activation of the innate immune system action of DMXAA is an indirect effect of DMXAA, a feature which is con- The structure of tumor vascularization is different from nor- sidered to be absent in other antivascular agents.[26] The mal tissues of the body.[17] Tumor vessels are immature be- induction of innate immune system stimulates inflamma- cause their hierarchical network build up is unlike normal tory cytokine production which is responsible for tumor- vascular tissues, in fact, they are unevenly distributed and specific inflammatory responses.[11,27] Approximately, chaotic.[5] Tumor endothelial cells are structurally and physi- 4 hr after injection of DMXAA, the amount of interleukin ologically abnormal, and more permeable to macromolecules 6 (IL- 6), tumor necrosis factor (TNF), granulocyte colony compared to normal tissues, and also their blood flow rate is stimulating factor (GCSF), and several chemokines—such far less than normal tissues.[18] These features are considered as macrophage inflammatory protein 1α (MIP-1α), IP- to be the major weaknesses of tumor vessels and, therefore, 10 and, KC—increases.[28–30] The rate of cytokine rising can be utilized as points of a target for eliminating tumors.[19] caused by DMXAA is greater in tumor tissues compared Two types of mechanisms of action have been proposed to plasma.[29] Although the molecular target of DMXAA for DMXAA: direct and indirect. DMXAA direct effect is yet unknown, it seems that, unlike the human stimu- lator of interferon gene (STING) that is not induced by DMXAA,[31] the induction of the innate immune system in mice occurs by STING. DMXAA stimulates pathways that have been observed in NFκB activation in mono- cytes,[30] vascular endothelial cells,[20] and various tumor cells.[32] NFκB also promotes the production of TNF and other inflammatory cytokines[33]—which change the or- ganization of the cytoskeleton in the tumor endothelial cells, thus, alerting their structure and shape.[34] DMXAA also escalates the levels of nitric oxide.[35] Both TNF and nitric oxide elicit vascular disrupting properties.[11] The production of TNF and nitric oxide along with other cyto- kines is the reason of tumor necrosis.[12] Three functions of increased permeability increased apoptosis, and de- creased blood flow is mediated by TNF because in mice lacking the TNF gene[36] or TNF receptor gene,[28] these three functions cannot be seen. The presence of TNF, how- ever, reduces the maximum tolerated dose of DMXAA. Consequently, TNF is an important factor affecting the performance and toxicity of DMXAA.[28] The other effects of DMXAA include activation of p38 (MAPK) and TBK1-IRF3 pathways,[37] and reduction of tumor energetics and membrane turnover.[38] Studies have, also, shown that DMXAA inhibits the activation of platelet and induces thrombosis through inhibition of phosphodies- [39] FIGURE 1 The chemical structure of DMXAA and related terase and thromboxane A2 signaling. Table 1 summa- compounds[12] rizes the proposed mechanisms for DMXAA. DAEI FARSHCHI ADLI ET AL. | 3 myelosuppression.[42,43] Significant clinical changes in vi- 1.3 Clinical efficacy | sion, retinal, and electrophysiological parameters were not The DMXAA drug can be utilized alone or in combination seen after six times injections of DMXAA.[44] The inves- with other anticancer agents to treat cancer. A web search tigation on the blood flow parameters made via a dynamic with the terms DMXAA, Vadimezan, and ASA404 in the contrast- enhanced MRI method on sixteen phase I patients database of clinical studies (https://clinicaltrials.gov) culmi- before and 6–24 hr after treatment with DMXAA showed nated in 18 results. These clinical trials are summarized in that DMXAA in doses of 500 to 4,900 had reduced the tumor Tables 2 and 3.
Recommended publications
  • Co-Administration of Vadimezan and Recombinant Coagulase-NGR Inhibits Growth of Melanoma Tumor in Mice
    Adv Pharm Bull, 2021, 11(2), 385-392 doi: 10.34172/apb.2021.037 TUOMS https://apb.tbzmed.ac.ir P R E S S Research Article Co-Administration of Vadimezan and Recombinant Coagulase-NGR Inhibits Growth of Melanoma Tumor in Mice Amir Daei Farshchi Adli1, Rana Jahanban-Esfahlan1, Khaled Seidi1, Davoud Farajzadeh2, Ramezan Behzadi3, Nosratollah Zarghami1,4* ID 1Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Table-fig Iran. 2Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran. 3North Research Center, Pasture Institute of Iran, Tehran, Amol, Iran. 4Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. Article info Abstract Article History: Purpose: Tumor vascular targeting appeared as an appealing approach to fight cancer, though, Received: 25 Jan. 2020 the results from the clinical trials and drugs in the market were proved otherwise. The promise of Revised: 1 Mar. 2020 anti-angiogenic therapy as the leading tumor vascular targeting strategy was negatively affected Accepted: 15 Apr. 2020 with the discovery that tumor vascularization can occur non-angiogenic mechanisms such as epublished: 15 Apr. 2020 co-option. An additional strategy is induction of tumor vascular infarction and ischemia. Methods: Such that we used truncated coagulase (tCoa) coupled to tumor endothelial targeting Keywords: moieties to produce tCoa-NGR fusion proteins. We showed that tCoa-NGR can bypass • DMXAA coagulation cascade to induce selective vascular thrombosis and infarction of mild and highly • Tumor vascular infarction proliferative solid tumors in mice.
    [Show full text]
  • DMXAA Causes Tumor Site-Specific Vascular Disruption
    DMXAA Causes Tumor Site-Specific Vascular Disruption in Murine Non-Small Cell Lung Cancer, and like the Endogenous Non-Canonical Cyclic Dinucleotide STING Agonist, 2939-cGAMP, Induces M2 Macrophage Repolarization Charlene M. Downey1,2, Mehrnoosh Aghaei1,2, Reto A. Schwendener3, Frank R. Jirik1,2* 1 Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada, 2 The McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada, 3 Institute of Molecular Cancer Research, Laboratory of Liposome Research, University of Zurich, Zurich, Switzerland Abstract The vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a murine agonist of the stimulator of interferon genes (STING), appears to target the tumor vasculature primarily as a result of stimulating pro-inflammatory cytokine production from tumor-associated macrophages (TAMs). Since there were relatively few reports of DMXAA effects in genetically-engineered mutant mice (GEMM), and models of non-small cell lung cancer (NSCLC) in particular, we examined both the effectiveness and macrophage dependence of DMXAA in various NSCLC models. The DMXAA responses of primary adenocarcinomas in K-rasLA1/+ transgenic mice, as well as syngeneic subcutaneous and metastatic tumors, generated by a p53R172HDg/+; K-rasLA1/+ NSCLC line (344SQ-ELuc), were assessed both by in vivo bioluminescence imaging as well as by histopathology. Macrophage-dependence of DMXAA effects was explored by clodronate liposome-mediated TAM depletion. Furthermore, a comparison of the vascular structure between subcutaneous tumors and metastases was carried out using micro-computed tomography (micro-CT). Interestingly, in contrast to the characteristic hemorrhagic necrosis produced by DMXAA in 344SQ-ELuc subcutaneous tumors, this agent failed to cause hemorrhagic necrosis of either 344SQ-ELuc-derived metastases or autochthonous K-rasLA1/+ NSCLCs.
    [Show full text]
  • Challenges and Opportunities in the Clinical Development of STING Agonists for Cancer Immunotherapy
    Journal of Clinical Medicine Review Challenges and Opportunities in the Clinical Development of STING Agonists for Cancer Immunotherapy Leila Motedayen Aval 1, James E. Pease 2 , Rohini Sharma 1 and David J. Pinato 1,* 1 Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W120HS, UK; [email protected] (L.M.A.); [email protected] (R.S.) 2 Inflammation, Repair & Development, National Heart & Lung Institute, Imperial College London, London SW7 2AZ, UK; [email protected] * Correspondence: [email protected]; Tel.: +44-(0)20-7594-2799 Received: 16 September 2020; Accepted: 12 October 2020; Published: 16 October 2020 Abstract: Immune checkpoint inhibitors (ICI) have revolutionised cancer therapy. However, they have been effective in only a small subset of patients and a principal mechanism underlying immune-refractoriness is a ‘cold’ tumour microenvironment, that is, lack of a T-cell-rich, spontaneously inflamed phenotype. As such, there is a demand to develop strategies to transform the tumour milieu of non-responsive patients to one supporting T-cell-based inflammation. The cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway is a fundamental regulator of innate immune sensing of cancer, with potential to enhance tumour rejection through the induction of a pro-inflammatory response dominated by Type I interferons. Recognition of these positive immune-modulatory properties has rapidly elevated the STING pathway as a putative target for immunotherapy, leading to a myriad of preclinical and clinical studies assessing natural and synthetic cyclic dinucleotides and non-nucleotidyl STING agonists. Despite pre-clinical evidence of efficacy, clinical translation has resulted into disappointingly modest efficacy.
    [Show full text]
  • Intratumoral STING Activation Sensitizes Poorly Immunogenic Cancers to Checkpoint Blockade Immunotherapy
    The Texas Medical Center Library DigitalCommons@TMC The University of Texas MD Anderson Cancer Center UTHealth Graduate School of The University of Texas MD Anderson Cancer Biomedical Sciences Dissertations and Theses Center UTHealth Graduate School of (Open Access) Biomedical Sciences 12-2018 Intratumoral STING Activation Sensitizes Poorly Immunogenic Cancers to Checkpoint Blockade Immunotherapy Casey Ager Follow this and additional works at: https://digitalcommons.library.tmc.edu/utgsbs_dissertations Part of the Medical Immunology Commons, and the Translational Medical Research Commons Recommended Citation Ager, Casey, "Intratumoral STING Activation Sensitizes Poorly Immunogenic Cancers to Checkpoint Blockade Immunotherapy" (2018). The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Dissertations and Theses (Open Access). 908. https://digitalcommons.library.tmc.edu/utgsbs_dissertations/908 This Dissertation (PhD) is brought to you for free and open access by the The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at DigitalCommons@TMC. It has been accepted for inclusion in The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Dissertations and Theses (Open Access) by an authorized administrator of DigitalCommons@TMC. For more information, please contact [email protected]. Approval Page INTRATUMORAL STING ACTIVATION SENSITIZES POORLY IMMUNOGENIC CANCERS TO CHECKPOINT BLOCKADE IMMUNOTHERAPY
    [Show full text]
  • Vascular Targeted Agents for the Treatment of Angiosarcoma
    Vascular Targeted Agents for the Treatment of Angiosarcoma Robin Joseph Young Thesis submitted for the degree of Doctor of Philosophy University of Sheffield Department of Oncology January 2013 Contents Section Title Page I Acknowledgements II Abstract III Abbreviations IV List of tables V List of figures 1 Introduction 1 1.1 Angiogenesis 1 1.1.1 Vascular Endothelial Growth Factors 2 1.1.2 Angiopoietins 3 1.1.3 Platelet Derived Growth Factors 4 1.1.4 Fibroblast Growth Factors 5 1.1.5 Notch 5 1.1.6 VEGF targeted therapy 6 1.1.6.1 Bevacizumab 6 1.1.6.2 Tyrosine kinase inhibitors 7 1.2 Angiosarcomas 11 1.2.1 Epidemiology 11 1.2.2 Aetiology 12 1.2.3 Clinical presentation 13 1.2.4 Pathology 14 1.2.5 Treatment 15 1.2.5.1 Localised disease 16 1.2.5.2 Advanced disease 16 1.2.5.3 Biological agents 17 1.3 The Molecular Pathology of Angiosarcomas 20 1.3.1 Chromosomal abnormalities 20 1.3.2 Immortality 21 1.3.3 Abnormal growth signals 21 1.3.4 Invasion and metastasis 23 1.3.5 Angiogenesis 24 1.4 Hypothesis 27 1.5 Aims 23 2 Methods 28 2.1 Cells 28 2.2 Characterisation Studies 31 2.2.1 Immunocytochemistry characterisation 31 2.2.1.1 Slides 31 2.2.1.2 Immunocytochemistry protocol 31 2.2.1.3 Permanox slides 32 2.2.2 Flow cytometry 33 2.2.3 Dil-Ac-LDL 35 2.3 Functional Assays 36 2.3.1 Growth kinetics 36 2.3.2 Differentiation 36 2.3.3 Migration 38 2.3.4 In vivo tumour formation 40 2.4 Protein Expression Profiling 40 2.4.1 Protein arrays 40 2.4.2 VEGF ELISA 43 2.4.3 Western blot 44 2.5 Drug Studies 49 2.5.1 Chemosensitivity assays 49 2.5.1.1 Cell membrane
    [Show full text]
  • WO 2018/217651 Al 29 November 2018 (29.11.2018) W !P O PCT
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2018/217651 Al 29 November 2018 (29.11.2018) W !P O PCT (51) International Patent Classification: KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, C07D 471/04 (2006.01) A61P 35/00 (2006.01) MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, A61K 31/519 (2006.01) C07D 475/00 (2006.01) OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY,TH, TJ, TM, TN, (21) International Application Number: TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. PCT/US2018/033714 (84) Designated States (unless otherwise indicated, for every (22) International Filing Date: kind of regional protection available): ARIPO (BW, GH, 2 1 May 2018 (21 .05.2018) GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, (25) Filing Language: English UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, (26) Publication Language: English EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, LV, (30) Priority Data: MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, 62/509,629 22 May 2017 (22.05.2017) US TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, GW, KM, ML, MR, NE, SN, TD, TG).
    [Show full text]
  • STING Agonist Inflames the Pancreatic Cancer Immune Microenvironment and Reduces Tumor Burden in Mouse Models Weiqing Jing1†, Donna Mcallister2†, Emily P
    Jing et al. Journal for ImmunoTherapy of Cancer (2019) 7:115 J Immunother Cancer: first published as 10.1186/s40425-019-0573-5 on 29 April 2019. Downloaded from https://doi.org/10.1186/s40425-019-0573-5 RESEARCHARTICLE Open Access STING agonist inflames the pancreatic cancer immune microenvironment and reduces tumor burden in mouse models Weiqing Jing1†, Donna McAllister2†, Emily P. Vonderhaar2, Katie Palen1,5, Matthew J. Riese1,2,3, Jill Gershan4, Bryon D. Johnson1,2,3,5† and Michael B. Dwinell2,3*† Abstract Pancreatic cancer is characterized by an immune suppressive stromal reaction that creates a barrier to therapy. A murine transgenic pancreatic cancer cell line that recapitulates human disease was used to test whether a STimulator of Interferon Genes (STING) agonist could reignite immunologically inert pancreatic tumors. STING agonist treatment potently changed the tumor architecture, altered the immune profile, and increased the survival of tumor-bearing mice. Notably, STING agonist increased numbers and activity of cytotoxic T cells within tumors and decreased levels of suppressive regulatory T cells. Further, STING agonist treatment upregulated costimulatory molecule expression on cross-presenting dendritic cells and reprogrammed immune-suppressive macrophages into immune-activating subtypes. STING agonist promoted the coordinated and differential cytokine production by dendritic cells, macrophages, and pancreatic cancer cells. Cumulatively, these data demonstrate that pancreatic cancer progression is potently inhibited by STING
    [Show full text]
  • Disrupting Tumour Vasculature and Recruitment of Apdl1-Loaded Platelets Control Tumour Metastasis
    ARTICLE https://doi.org/10.1038/s41467-021-22674-3 OPEN Disrupting tumour vasculature and recruitment of aPDL1-loaded platelets control tumour metastasis Hongjun Li1,2,3,4,5, Zejun Wang 3,4,5, Zhaowei Chen1,3,6, Tianyuan Ci3, Guojun Chen3,4,5, Di Wen3,4,5, Ruoxin Li3,5, Jinqiang Wang1,3,4,5, Huan Meng 5, R. Bryan Bell 7, Zhifeng Gu 8, Gianpietro Dotti 9 & ✉ Zhen Gu 1,2,3,4,5 Although therapies of cancer are advancing, it remains challenging for therapeutics to reach 1234567890():,; the sites of metastasis, which accounts for majority of cancer associated death. In this study, we have developed a strategy that guides an anti-programmed cell death-ligand 1 (aPDL1) antibody to accumulate in metastatic lesions to promote anti-tumour immune responses. Briefly, we have developed a combination in which Vadimezan disrupts tumour blood vessels of tumour metastases and facilitates the recruitment and activation of adoptively transferred aPDL1-conjugated platelets. In situ activated platelets generate aPDL1-decorated platelet- derived microparticles (PMP) that diffuse within the tumour and elicit immune responses. The proposed combination increases 10-fold aPDL1 antibody accumulation in lung metas- tases as compared to the intravenous administration of the antibody and enhances the magnitude of immune responses leading to improved antitumour effects. 1 College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China. 2 Zhejiang University Medical Center, Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, China. 3 Department of Bioengineering, University of California, Los Angeles, CA, USA. 4 Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
    [Show full text]
  • WO 2021/026099 A1 11 February 2021 (11.02.2021) W P O PCT
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization II III International Bureau (10) International Publication Number (43) International Publication Date WO 2021/026099 A1 11 February 2021 (11.02.2021) W P O PCT (51) International Patent Classification: NZ, OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, C07D 241/28 (2006.01) A61P 35/00 (2006.01) SA, SC, SD, SE, SG, SK, SL, ST, SV, SY, TH, TJ, TM, TN, C07D 401/04 (2006.01) A61K 31/497 (2006.01) TR, TT, TZ, UA, UG, US, UZ, VC, VN, WS, ZA, ZM, ZW. C07D 401/14 (2006.01) (84) Designated States (unless otherwise indicated, for every (21) International Application Number: kind of regional protection available) . ARIPO (BW, GH, PCT/US2020/044798 GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, (22) International Filing Date: TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, 03 August 2020 (03.08.2020) EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, LV, (25) Filing Language: English MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, TR), OAPI (BF, BJ, CF, CG, Cl, CM, GA, GN, GQ, GW, (26) Publication Language: English KM, ML, MR, NE, SN, TD, TG). (30) Priority Data: 62/882,265 02 August 2019 (02.08.2019) US Declarations under Rule 4.17: — as to applicant's entitlement to apply for and be granted a (71) Applicant: AMGEN INC.
    [Show full text]
  • Discovery of Potential Therapeutic Targets for Non-Small Cell Lung
    RSC Advances View Article Online PAPER View Journal | View Issue Discovery of potential therapeutic targets for non- small cell lung cancer using high-throughput Cite this: RSC Adv.,2019,9, 10905 metabolomics analysis based on liquid chromatography coupled with tandem mass spectrometry† Hong-dan Xu,a Wen Luo,b Yuanlong Lin,c Jiawen Zhang,b Lijuan Zhang,b Wei Zhang *b and Shu-ming Huang*d Lung cancer is a severe health problem and threatens a patient's quality of life. The metabolites present in biological systems are expected to be key mediators and the changes in these metabolites play an important role in promoting health. Metabolomics can unravel the global metabolic changes and identify significant biological pathways involved in disease development. However, the role of metabolites in lung cancer is still Creative Commons Attribution-NonCommercial 3.0 Unported Licence. largely unknown. In the present study, we developed a liquid chromatography coupled with tandem mass spectrometry method for biomarker discovery and identification of non-small cell lung cancer (NSCLC) from metabolomics data sets and aimed to investigate the metabolic profiles of NSCLC samples to identify potential disease biomarkers and to reveal the pathological mechanism. After cell metabolite extraction, the metabolic changes in NSCLC cells were characterized and targeted metabolite analysis was adopted to offer a novel opportunity to probe into the relationship between differentially regulated cell metabolites and NSCLC. Quantitative analysis of key enzymes in the disturbed pathways by proteomics was employed to verify metabolomic pathway changes. A total of 13 specific biomarkers were identified in NSCLC cells related with metabolic disturbance of NSCLC morbidity, which were involved in 4 vital pathways, namely glycine, serine and This article is licensed under a threonine metabolism, aminoacyl-tRNA biosynthesis, tyrosine metabolism and sphingolipid metabolism.
    [Show full text]
  • Prostate-Specific Antigen-Based Screening for Prostate Cancer: an Evidence Update for the U.S
    Evidence Synthesis_______________________________ Number 90 Prostate-Specific Antigen-Based Screening for Prostate Cancer: An Evidence Update for the U.S. Preventive Services Task Force Prepared for: Agency for Healthcare Research and Quality U.S. Department of Health and Human Services 540 Gaither Road Rockville, MD 20850 www.ahrq.gov Prepared by: Kenneth Lin, MD Jennifer M. Croswell, MD, MPH Helen Koenig, MD, MPH Clarence Lam, MD, MPH Ashley Maltz, MD, MPH AHRQ Publication No. 12-05160-EF-1 October 2011 This report is based on research conducted by staff of the Agency for Healthcare Research and Quality (AHRQ), Rockville, MD. No statement in this report should be construed as an official position of AHRQ or of the U.S. Department of Health and Human Services. The information in this report is intended to help clinicians, employers, policymakers, and others make informed decisions about the provision of health care services. This report is intended as a reference and not as a substitute for clinical judgment. This report may be used, in whole or in part, as the basis for the development of clinical practice guidelines and other quality enhancement tools, or as a basis for reimbursement and coverage policies. AHRQ or U.S. Department of Health and Human Services endorsement of such derivative products may not be stated or implied. Suggested Citation: Lin K, Croswell JM, Koenig H, Lam C, Maltz A. Prostate-Specific Antigen-Based Screening for Prostate Cancer: An Evidence Update for the U.S. Preventive Services Task Force. Evidence Synthesis No. 90. AHRQ Publication No. 12-05160-EF-1.
    [Show full text]
  • Sponsor Novartis Generic Drug Name Vadimezan Therapeutic Area
    Clinical Trial Results Database Page 1 Sponsor Novartis Generic Drug Name Vadimezan Therapeutic Area of Trial Non-small-cell lung cancer (NSCLC) Approved Indication Investigational Study Number CASA404A2302 Title A phase III, randomized, double-blind, placebo-controlled, multi-center study of Vadimezan in combination with docetaxel in second-line treatment of patients with locally advanced or metastatic (stage III b/IV) non-small-cell lung cancer (NSCLC) Phase of Development Phase III Study Start/End Dates 19 Dec 2008 to 20 Dec 2010 Study Design/Methodology This was a prospective, global, multi-center, double-blind, placebo-controlled, randomized Phase III trial. Patients were randomized in a 1:1 ratio into one of the following two treatment arms ASA404 1800 mg/m2 plus docetaxel 75 mg/m2 or to placebo plus docetaxel 75 mg/m2. Randomization was stratified by: World health Organization (WHO) performance status 0-1 versus 2; histology (squamous versus non-squamous); prior treatment with a paclitaxel-based regimen, if any, in the first-line setting. Centres 218 centres in 23 countries: Argentina (5), Belgium (8), Brazil (13), Canada (10), China (9), Egypt (2), Germany (21), Hungary (7), Italy (12), Japan (15), Lebanon (1), Luxembourg (1), Mexico (1), Netherlands (3), New Zealand (4), Poland (4), South Africa (3)Spain (4), Switzerland (2), Thailand (3), Turkey (4), UK (8), USA (78) Clinical Trial Results Database Page 2 Publication: None. Objectives Primary objective(s) To compare the Overall survival (OS) between the Vadimezan plus docetaxel group and the placebo plus docetaxel group. Secondary objective(s) Key secondary objective: To compare Progression-free Survival (PFS) and the Overall Response Rate (ORR) per Response Evaluation Criteria in Solid Tumors(RECIST) assessed by the investigator between patients receiving Vadimezan or placebo in combination with docetaxel.
    [Show full text]