Multiscale analysis of metabolic inflammation as a driver of breast cancer
代谢炎症作为乳腺癌驱动因素的多尺度分析
基本信息
- 批准号:10259753
- 负责人:
- 金额:$ 56.1万
- 依托单位:
- 依托单位国家:美国
- 项目类别:
- 财政年份:2020
- 资助国家:美国
- 起止时间:2020-09-09 至 2025-08-31
- 项目状态:未结题
- 来源:
- 关键词:AcuteAddressAdipose tissueAffectAlgorithmsAmericanApoptoticAutomobile DrivingBase SequenceBiological AssayBlocking AntibodiesBreastBreast Cancer CellBreast Cancer ModelBreast Cancer PatientBromodomainCD8B1 geneCancer BurdenCancer ModelCancer PatientCell modelCellsCellular Metabolic ProcessChemicalsChronicCitiesClinicalCoculture TechniquesComplexComputer ModelsDangerousnessDataDevelopmentDiabetes MellitusDiseaseEndocrinologyEstrogen receptor negativeGenesGoalsHospitalsHypertensionImmuneImmunologyImmunooncologyImmunophenotypingImmunotherapyInflammationInflammatoryKnowledgeMalignant NeoplasmsMammary NeoplasmsMass Spectrum AnalysisMedical OncologyMetabolicMetabolic DiseasesMetabolismMetforminMicrometastasisModelingMolecularNeoplasm MetastasisNewly DiagnosedNon-Insulin-Dependent Diabetes MellitusObesityOrganoidsOutcomePathway AnalysisPathway interactionsPatient-Focused OutcomesPatientsPharmaceutical PreparationsPopulation SciencesPrediabetes syndromePrevalencePrimary NeoplasmProcessPrognosisProspective StudiesProteinsProteomePublic HealthPublishingResearch PersonnelResolutionRiskSignal TransductionSuggestionSurgical OncologySystemSystems BiologyT cell receptor repertoire sequencingT-LymphocyteT-Lymphocyte SubsetsTalentsTestingThe Cancer Genome AtlasTumor ImmunityTumor-DerivedTumor-infiltrating immune cellsWomanbasebreast cancer progressioncancer subtypescell killingcheckpoint receptorschronic inflammatory diseaseclinically relevantcohortcomorbidityepidemiologic dataexhaustexhaustionexperienceexperimental studyimmune checkpointimprovedinhibitor/antagonistinnovationmalignant breast neoplasmmathematical modelmetabolic profilemortalitymulti-scale modelingmultidisciplinarymultiple omicsneoplasm immunotherapyneoplastic cellnetwork modelsnon-diabeticnovelpatient populationphosphoproteomicspolarized cellprimary outcomeprofiles in patientsresponsesafety nettranscriptometranscriptome sequencingtreatment responsetriple-negative invasive breast carcinomatumortumor microenvironmenttumor progression
项目摘要
Women with breast cancer and co-morbid Type 2 diabetes (T2D) have up to 40% worse overall survival
compared to non-diabetic women; this co-morbidity burden is disproportionately high among vulnerable cohorts,
such as patients at safety net hospitals in the U.S., where it can affect half of the patient population. Yet, current
models of breast tumor progression and immunotherapy are based on data from metabolically healthy cancer
patients, ignoring metabolic /inflammatory components of T2D. Preliminary and published data support an overall
hypothesis: specific metabolic and immune exhaustion networks in breast cancer patients with co-morbid T2D
promote tumor aggressiveness. We propose an innovative multiscale modelling framework to identify these
networks by integrating metabolic, inflammatory and immune signatures in multi-omics cancer models
encompassing RNA-seq and phosphoproteomics data. We take a systems biology approach to combine
innovative computational, clinical and patient-derived tumor organoid experiments to investigate interactions
among putative driver genes, T2D and immune exhaustion, with tumor progression/aggressiveness as the
primary outcome variable in estrogen receptor-negative (ER-) breast cancer, which has poor prognosis and is
highly prevalent among safety net hospital patients. We will model how T2D rewires signaling hubs, nodes and
edges in newly diagnosed breast cancer patients, then test these networks in breast organoid models. We will
develop a unified model through three Aims: Aim 1: Determine how T2D reprograms immune exhaustion and
metabolism in the tumor microenvironment of ER negative (ER-) breast cancer. We will apply RNAseq and
scRNAseq to primary ER- breast cancer cells and tumor immune infiltrates to compare three groups of patients
(T2D, T2D+ metformin-medicated (T2D+M), non-diabetic (ND) controls) to construct a preliminary network
supplemented with TCGA data. Differential gene and pathway analyses will elucidate regulatory relationships
and key hubs. We hypothesize that the connectivity of the ER- cluster in T2D will be altered and denser than in
ND or T2D+M. Aim 2: We will generate patient-derived organoids, including organoid-primed T cells (OpT), to
test the computational model for metabolism and immune checkpoints. We will evaluate mechanistic hypotheses
that T2D medications, immune checkpoint-blocking antibodies and chemical inhibitors of BET bromodomain
proteins (which regulate checkpoint expression) overcome immune exhaustion to improve OpT cell metabolism
and tumor cell killing. TCR sequencing will reveal emergent OpT oligoclonality; deep immunophenotyping will
reveal T2D-driven signaling networks. Aim 3: Determine abnormal signaling networks impacting cancer immunity
in organoid and OpT models. We will perform deep phosphoproteomic profiling of primary tumors, organoids,
circulating T cells and OpT cells, from the three metabolic groups, then use pathway projection and network
analyses to refine our integrated model. Together, our unique systems biology approach will capture the complex
interactions among tumor, immune infiltrates and metabolic genes to address the cancer burden of T2D.
患有乳腺癌和合并2型糖尿病(T2 D)的女性的总生存率最多下降40%
与非糖尿病妇女相比;这种合并症负担在脆弱群体中不成比例地高,
比如美国安全网医院的病人,它可以影响一半的患者群体。然而,目前
乳腺肿瘤进展和免疫治疗的模型是基于代谢健康癌症的数据
患者,忽略T2 D的代谢/炎症成分。初步和公布的数据支持总体
假设:乳腺癌合并T2 D患者的特异性代谢和免疫耗竭网络
促进肿瘤的侵袭性。我们提出了一个创新的多尺度建模框架,以确定这些
通过在多组学癌症模型中整合代谢、炎症和免疫特征,
包括RNA-seq和磷酸蛋白质组学数据。我们采用系统生物学方法将联合收割机
创新的计算,临床和患者来源的肿瘤类器官实验,以研究相互作用
在假定的驱动基因中,T2 D和免疫衰竭,肿瘤进展/侵袭性是
雌激素受体阴性(ER-)乳腺癌的主要结局变量,预后不良,
在安全网医院的病人中非常普遍。我们将对T2 D如何重新连接信令集线器、节点和
边缘,然后在乳腺类器官模型中测试这些网络。我们将
目标1:确定T2 D如何重新编程免疫衰竭,
在ER阴性(ER-)乳腺癌的肿瘤微环境中的代谢。我们将应用RNAseq,
scRNAseq对原发性ER-乳腺癌细胞和肿瘤免疫浸润进行比较三组患者
(T2D、T2 D+二甲双胍给药(T2 D +M)、非糖尿病(ND)对照)以构建初步网络
并补充了TCGA数据。差异基因和途径分析将阐明调控关系
和关键枢纽。我们假设T2 D中ER-簇的连接性将比T2 D中的ER-簇的连接性改变且更密集。
ND或T2 D +M。目标2:我们将产生患者源性类器官,包括类器官致敏T细胞(OpT),
测试代谢和免疫检查点的计算模型。我们将评估机械假说
T2 D药物、免疫检查点阻断抗体和BET布罗莫结构域的化学抑制剂
蛋白质(调节检查点表达)克服免疫耗竭,改善OpT细胞代谢
和肿瘤细胞杀伤。TCR测序将揭示出现的OpT寡克隆性;深度免疫表型将
揭示T2 D驱动的信号网络。目的3:确定影响癌症免疫力的异常信号网络
在类器官和OpT模型中。我们将对原发性肿瘤,类器官,
循环T细胞和OpT细胞,从三个代谢组,然后使用通路投射和网络
分析以完善我们的综合模型。总之,我们独特的系统生物学方法将捕捉复杂的
肿瘤,免疫浸润和代谢基因之间的相互作用,以解决T2 D的癌症负担。
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 }}
Gerald V Denis其他文献
Gerald V Denis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('Gerald V Denis', 18)}}的其他基金
Multiscale analysis of metabolic inflammation as a driver of breast cancer
代谢炎症作为乳腺癌驱动因素的多尺度分析
- 批准号:
10063646 - 财政年份:2020
- 资助金额:
$ 56.1万 - 项目类别:
Multiscale analysis of metabolic inflammation as a driver of breast cancer
代谢炎症作为乳腺癌驱动因素的多尺度分析
- 批准号:
10473886 - 财政年份:2020
- 资助金额:
$ 56.1万 - 项目类别:
Mechanisms of BET bromodomain metabolic reprogramming in triple negative breast cancer
三阴性乳腺癌中 BET 溴结构域代谢重编程的机制
- 批准号:
10217042 - 财政年份:2018
- 资助金额:
$ 56.1万 - 项目类别:
Mechanisms of BET bromodomain metabolic reprogramming in triple negative breast cancer
三阴性乳腺癌中 BET 溴结构域代谢重编程的机制
- 批准号:
10442588 - 财政年份:2018
- 资助金额:
$ 56.1万 - 项目类别:
Mechanisms of BET bromodomain metabolic reprogramming in triple negative breast cancer
三阴性乳腺癌中 BET 溴结构域代谢重编程的机制
- 批准号:
9757730 - 财政年份:2018
- 资助金额:
$ 56.1万 - 项目类别:
Uncoupling obesity from breast cancer in African American women
非洲裔美国女性肥胖与乳腺癌的关系
- 批准号:
9337393 - 财政年份:2013
- 资助金额:
$ 56.1万 - 项目类别:
Uncoupling obesity from breast cancer in African American women
非洲裔美国女性肥胖与乳腺癌的关系
- 批准号:
8633292 - 财政年份:2013
- 资助金额:
$ 56.1万 - 项目类别:
Uncoupling obesity from breast cancer in African American women
非洲裔美国女性肥胖与乳腺癌的关系
- 批准号:
8740475 - 财政年份:2013
- 资助金额:
$ 56.1万 - 项目类别:
Uncoupling obesity from breast cancer in African American women
非洲裔美国女性肥胖与乳腺癌的关系
- 批准号:
9134718 - 财政年份:2013
- 资助金额:
$ 56.1万 - 项目类别:
Mechanisms of Brd2 immunoprotection from insulin resistance
Brd2 免疫保护胰岛素抵抗的机制
- 批准号:
8332905 - 财政年份:2011
- 资助金额:
$ 56.1万 - 项目类别:
相似海外基金
Rational design of rapidly translatable, highly antigenic and novel recombinant immunogens to address deficiencies of current snakebite treatments
合理设计可快速翻译、高抗原性和新型重组免疫原,以解决当前蛇咬伤治疗的缺陷
- 批准号:
MR/S03398X/2 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Fellowship
Re-thinking drug nanocrystals as highly loaded vectors to address key unmet therapeutic challenges
重新思考药物纳米晶体作为高负载载体以解决关键的未满足的治疗挑战
- 批准号:
EP/Y001486/1 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Research Grant
CAREER: FEAST (Food Ecosystems And circularity for Sustainable Transformation) framework to address Hidden Hunger
职业:FEAST(食品生态系统和可持续转型循环)框架解决隐性饥饿
- 批准号:
2338423 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Continuing Grant
Metrology to address ion suppression in multimodal mass spectrometry imaging with application in oncology
计量学解决多模态质谱成像中的离子抑制问题及其在肿瘤学中的应用
- 批准号:
MR/X03657X/1 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Fellowship
CRII: SHF: A Novel Address Translation Architecture for Virtualized Clouds
CRII:SHF:一种用于虚拟化云的新型地址转换架构
- 批准号:
2348066 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Standard Grant
The Abundance Project: Enhancing Cultural & Green Inclusion in Social Prescribing in Southwest London to Address Ethnic Inequalities in Mental Health
丰富项目:增强文化
- 批准号:
AH/Z505481/1 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Research Grant
ERAMET - Ecosystem for rapid adoption of modelling and simulation METhods to address regulatory needs in the development of orphan and paediatric medicines
ERAMET - 快速采用建模和模拟方法的生态系统,以满足孤儿药和儿科药物开发中的监管需求
- 批准号:
10107647 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
EU-Funded
BIORETS: Convergence Research Experiences for Teachers in Synthetic and Systems Biology to Address Challenges in Food, Health, Energy, and Environment
BIORETS:合成和系统生物学教师的融合研究经验,以应对食品、健康、能源和环境方面的挑战
- 批准号:
2341402 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Standard Grant
Ecosystem for rapid adoption of modelling and simulation METhods to address regulatory needs in the development of orphan and paediatric medicines
快速采用建模和模拟方法的生态系统,以满足孤儿药和儿科药物开发中的监管需求
- 批准号:
10106221 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
EU-Funded
Recite: Building Research by Communities to Address Inequities through Expression
背诵:社区开展研究,通过表达解决不平等问题
- 批准号:
AH/Z505341/1 - 财政年份:2024
- 资助金额:
$ 56.1万 - 项目类别:
Research Grant