Development of (R,S')-MNF as a dual-targeted therapy for pancreatic cancer
(R,S)-MNF作为胰腺癌双靶向疗法的开发
基本信息
- 批准号:10546773
- 负责人:
- 金额:$ 38.51万
- 依托单位:
- 依托单位国家:美国
- 项目类别:
- 财政年份:2022
- 资助国家:美国
- 起止时间:2022-09-01 至 2024-08-31
- 项目状态:已结题
- 来源:
- 关键词:ABCB1 geneABCB6 geneAdenylate CyclaseAftercareAgonistAnabolismAnimalsAntineoplastic AgentsAttenuatedAutophagocytosisBiodistributionBiologicalBiological AssayBiological MarkersBloodCA-19-9 AntigenCancer EtiologyCell LineCellsCessation of lifeCyclic AMP-Dependent Protein KinasesDataDevelopmentDoseDown-RegulationDrug KineticsDrug TargetingDrug usageEnzyme-Linked Immunosorbent AssayEvaluationFormulationG-Protein-Coupled ReceptorsGPR55 receptorGTP-Binding Protein alpha Subunits, GsGenesGenetic TranscriptionGlutamate Metabolism PathwayGlycolysisGoalsHumanImmunoblot AnalysisImmunohistochemistryKnowledgeLiquid ChromatographyLysophosphatidylcholinesMEKsMalignant neoplasm of pancreasMass Spectrum AnalysisMaximum Tolerated DoseMetabolicMetabolismModelingMonitorMusOrganOxidative StressPI3K/AKTPaclitaxelPancreatic Ductal AdenocarcinomaPatientsPharmaceutical PreparationsPharmacodynamicsPharmacologic SubstancePharmacotherapyPhasePlasmaPredictive ValuePreparationProcessPrognosisProteinsProtocols documentationPyrimidineRegimenResistanceSamplingScheduleSignal TransductionSmall Business Technology Transfer ResearchSurvival RateTechniquesTherapeuticTherapeutic AgentsTherapeutic UsesTissuesToxic effectTransforming Growth Factor betaTreatment ProtocolsTumor TissueXenograft Modelattenuationbasebeta cateninbeta-2 Adrenergic Receptorsc-myc Genescancer therapycell growthchemotherapyexperiencegemcitabineglucose metabolismimprovedinhibitorlipid metabolismmetabolomicsmouse modelnovel therapeuticspancreatic cancer patientspancreatic ductal adenocarcinoma cellpancreatic ductal adenocarcinoma modelpatient derived xenograft modelpharmacokinetics and pharmacodynamicsphase 1 studyprotein biomarkersresponse biomarkersmall moleculestandard of caretargeted treatmenttherapeutic evaluationtherapeutic targettranscription factortranscriptomicstreatment grouptumortumor growthtumor xenograft
项目摘要
Summary
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the U.S.
with a 5-year survival rate of <9%. The poor prognosis is partially due to resistance to standard
of care treatments including gemcitabine (Gem) and Gem+nab-paclitaxel (n-PTX). The
transcription factors HIF-1α and c-MYC are at the center of the mechanisms producing Gem and
n-PTX resistance and are key therapeutic targets. (R,S′)-4′-Methoxy-1-naphthylfenoterol (R,S’)-
MNF is a bi-functional anti-cancer agent that acts as a competitive inhibitor of GPR55 and a
biased-agonist of the β2-adrenergic receptor. In a PANC-1 xenograft tumor model (R,S’)-MNF
significantly dampens tumor growth, ∼75% (p<0.01), and downregulates HIF-1α and c-MYC
expression. Our overarching hypothesis is that (R,S′)-MNF will reduce PDAC tumor growth as
a single agent and produce positive synergistic effects with standard of care agents. The overall
goal is to determine the therapeutic potential of (R,S′)-MNF in combination with GEM+n-PTX in
PDAC models. The experimental protocols will utilize our knowledge of (R,S′)-MNF
pharmacokinetics and toxicity and experience with use of therapeutic agents in PDAC models.
Specific aims are: Aim 1: to determine the antitumor activity of (R,S′)-MNF alone and in
combination in PDAC patient derived xenograft (PDX) models: The initial step will be a dosing
finding study to determine maximal tolerated dose of (R,S′)-MNF alone and in combination with
GEM (70 mg/kg, i.p., once a week for 3 weeks) + n-PTX (30 mg/kg, once a week for 3 weeks).
Optimal dose and schedule will be used in 2 PDX models derived from PDAC patients’ tumors
expressing high levels of GPR55 and β2-AR. Each study will include 4 treatment groups of 18
mice/group: vehicle, (R,S′)-MNF alone, GEM+n-PTX, and (R,S′)-MNF + GEM+n-PTX. Blood and
major organ tissues will be collected from 6 mice/group for analyses proposed in Aim 2. The
remaining mice (12/group) will be monitored to determine the effect on tumor growth and survival.
Aim 2: to identify treatment biomarkers and determine (R,S′)-MNF biodistributions: Plasma
samples collected before and after treatment will be analyzed using LC-MS/MS to quantify small
molecules such as lysophosphatidylcholines (14:0 and16:0) and lactate and ELISA assays for
PDAC biomarkers such as CA19-9 and CYR61. Metabolite and protein concentrations will be
compared to tumor growth and survival data and to the relative expression of GPR55 and β2-AR.
Major organs collected from the (R,S′)-MNF treatment group will be analyzed using LC-MS/MS to
determine drug biodistribution. Data from the Phase I study will support IND studies in a Phase
II application, which will include GLP PK/PD, metabolism and toxicity studies.
概括
胰腺导管腺癌 (PDAC) 是美国癌症死亡的第三大原因。
5年生存率<9%,预后不良部分是由于对标准的抵抗。
护理治疗包括吉西他滨 (Gem) 和 Gem+白蛋白结合型紫杉醇 (n-PTX)。
转录因子 HIF-1α 和 c-MYC 是产生 Gem 和
n-PTX 耐药性是关键的治疗靶点 (R,S')-4'-甲氧基-1-萘非特罗 (R,S')-。
MNF 是一种双功能抗癌剂,可作为 GPR55 的竞争性抑制剂和
PANC-1 异种移植肿瘤模型 (R,S’)-MNF 中的 β2-肾上腺素受体偏向激动剂。
显着抑制肿瘤生长,约 75% (p<0.01),并下调 HIF-1α 和 c-MYC
我们的总体假设是 (R,S')-MNF 将减少 PDAC 肿瘤的生长
单一药物并与标准护理药物产生积极的协同效应。
目标是确定 (R,S')-MNF 与 GEM+n-PTX 组合的治疗潜力
PDAC 模型将利用我们对 (R,S')-MNF 的了解。
药代动力学和毒性以及在 PDAC 模型中使用治疗药物的经验。
具体目标是: 目标 1:确定 (R,S')-MNF 单独和单独使用的抗肿瘤活性
PDAC 患者衍生异种移植 (PDX) 模型中的组合:第一步是给药
发现研究确定 (R,S')-MNF 单独使用和联合使用的最大耐受剂量
GEM(70 mg/kg,腹腔注射,每周一次,持续 3 周)+ n-PTX(30 mg/kg,每周一次,持续 3 周)。
最佳剂量和方案将用于源自 PDAC 患者肿瘤的 2 个 PDX 模型
表达高水平的 GPR55 和 β2-AR 每项研究将包括 4 个治疗组,每组 18 人。
小鼠/组:载体、单独的(R,S')-MNF、GEM+n-PTX和(R,S')-MNF+GEM+n-PTX血液和。
将从每组 6 只小鼠收集主要器官组织,用于目标 2 中提出的分析。
将监测剩余的小鼠(12只/组)以确定对肿瘤生长和存活的影响。
目标 2:识别治疗生物标志物并确定 (R,S')-MNF 生物分布:血浆
治疗前后收集的样本将使用 LC-MS/MS 进行分析,以量化小
溶血磷脂酰胆碱(14:0 和 16:0)和乳酸等分子以及 ELISA 检测
PDAC 生物标志物如 CA19-9 和 CYR61 代谢物和蛋白质浓度。
与肿瘤生长和存活数据以及 GPR55 和 β2-AR 的相对表达进行比较。
从 (R,S')-MNF 治疗组收集的主要器官将使用 LC-MS/MS 进行分析,以
确定药物的生物分布。第一阶段研究的数据将支持第二阶段的 IND 研究。
II申请,将包括GLP PK/PD、代谢和毒性研究。
项目成果
期刊论文数量(0)
专著数量(0)
科研奖励数量(0)
会议论文数量(0)
专利数量(0)
数据更新时间:{{ journalArticles.updateTime }}
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
数据更新时间:{{ journalArticles.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ monograph.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ sciAawards.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ conferencePapers.updateTime }}
{{ item.title }}
- 作者:
{{ item.author }}
数据更新时间:{{ patent.updateTime }}
Haiyong Han其他文献
Haiyong Han的其他文献
{{
item.title }}
{{ item.translation_title }}
- DOI:
{{ item.doi }} - 发表时间:
{{ item.publish_year }} - 期刊:
- 影响因子:{{ item.factor }}
- 作者:
{{ item.authors }} - 通讯作者:
{{ item.author }}
{{ truncateString('Haiyong Han', 18)}}的其他基金
Molecular mechanisms of perineural invasion in pancreatic cancer
胰腺癌神经周围浸润的分子机制
- 批准号:
7707218 - 财政年份:2009
- 资助金额:
$ 38.51万 - 项目类别:
相似国自然基金
ABCB6基因在眼组织缺损中的功能研究
- 批准号:81470665
- 批准年份:2014
- 资助金额:80.0 万元
- 项目类别:面上项目
遗传性泛发性色素异常症致病基因及相关致病机理的研究
- 批准号:81371723
- 批准年份:2013
- 资助金额:51.0 万元
- 项目类别:面上项目
ABCB6基因在眼缺损发病机制中的功能研究
- 批准号:81371061
- 批准年份:2013
- 资助金额:70.0 万元
- 项目类别:面上项目
色素代谢新基因ABCB6的功能研究
- 批准号:31171228
- 批准年份:2011
- 资助金额:60.0 万元
- 项目类别:面上项目
相似海外基金
Doxorubicin cardiotoxicity and the protective effects of exercise
阿霉素心脏毒性和运动的保护作用
- 批准号:
10162657 - 财政年份:2019
- 资助金额:
$ 38.51万 - 项目类别:
Doxorubicin-induced respiratory dysfunction and the protective effects of exercise
阿霉素引起的呼吸功能障碍及运动的保护作用
- 批准号:
10449081 - 财政年份:2019
- 资助金额:
$ 38.51万 - 项目类别:
Doxorubicin cardiotoxicity and the protective effects of exercise
阿霉素心脏毒性和运动的保护作用
- 批准号:
10594901 - 财政年份:2019
- 资助金额:
$ 38.51万 - 项目类别:
Doxorubicin cardiotoxicity and the protective effects of exercise
阿霉素心脏毒性和运动的保护作用
- 批准号:
9897610 - 财政年份:2019
- 资助金额:
$ 38.51万 - 项目类别:
Doxorubicin cardiotoxicity and the protective effects of exercise
阿霉素心脏毒性和运动的保护作用
- 批准号:
10386915 - 财政年份:2019
- 资助金额:
$ 38.51万 - 项目类别: