Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT2-10-01
A. Cheng, M. Levy
Patients with breast cancer spend significant time1, effort, and financial resources2 to combat the disease for years after their diagnosis. The large volume of healthcare tasks can cause patients to become overburdened, leading to reduced adherence with care plans and worse outcomes3. On the other hand, certain patient characteristics such as physical resilience, financial well-being, and supportive family environments increase patients9 capacity to manage care4. Assessing treatment burden and capacity when prescribing care has been applied to populations such as diabetes patients5. We are investigating this paradigm in treatment of patients with breast cancer. The goal of this preliminary study is to identify significant factors that contribute to treatment burden, capacity to manage care, and outcomes of overburden for patients with breast cancer. Through literature review, interviews with survivors, and expert panels of navigators and providers, we will develop a survey instrument given to patients at the time of diagnosis. The survey will assess patient capacity and help providers give treatment options based on attributes of the patient. Additionally, we will attempt to correlate survey results with treatment burden measures derived from electronic health record data at a population level1. With treatment personalized for patient capacity, patients should be better able to adhere to care plans leading to improved quality of life during treatment and beyond. Acknowledgements: The authors would like to thank Cheryl Jernigan, our patient advocate mentor, for her guidance in this project. We would also like to thank the Susan G. Komen Foundation for their support of this research. References: 1. Cheng, A. C. & Levy, M. A. Data Driven Approach to Burden of Treatment Measurement: A Study of Patients with Breast Cancer. AMIA Annu. Symp. proceedings. AMIA Symp.2016, 1756–1763 (2016). 2. Zafar, S. Y. et al. The financial toxicity of cancer treatment: a pilot study assessing out-of-pocket expenses and the insured cancer patient9s experience. Oncologist.18, 381–90 (2013). 3. Mair, F. S. & May, C. R. Thinking about the burden of treatment. BMJ.349, g6680–g6680 (2014). 4. Boehmer, K. R., Shippee, N. D., Beebe, T. J. & Montori, V. M. Pursuing Minimally Disruptive Medicine: Correlation of patient capacity with disruption from illness and healthcare-related demands. J. Clin. Epidemiol. (2016). 5. Ishii, H. et al. Reproducibility and Validity of a Questionnaire Measuring Treatment Burden on Patients with Type 2 Diabetes: Diabetic Treatment Burden Questionnaire (DTBQ). Diabetes Ther.9, 1001–1019 (2018). Citation Format: Cheng A, Levy M. Treatment burden and capacity to manage care among patients with breast cancer [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT2-10-01.
乳腺癌患者在确诊后的数年里都要花费大量的时间、精力和财力与疾病作斗争。大量的医疗保健任务可能导致患者负担过重,从而降低对护理计划的依从性,并导致更糟糕的结果。另一方面,病人的某些特征,如身体恢复能力、经济状况良好和支持性的家庭环境,提高了病人管理护理的能力。在对糖尿病患者等人群实施处方护理时,评估治疗负担和能力5。我们正在研究这种治疗乳腺癌患者的模式。本初步研究的目的是确定影响乳腺癌患者治疗负担、护理管理能力和过度负担结果的重要因素。通过文献回顾,与幸存者的访谈,以及由导航员和提供者组成的专家小组,我们将开发一种在诊断时给予患者的调查工具。该调查将评估患者的能力,并帮助提供者根据患者的属性提供治疗方案。此外,我们将尝试将调查结果与从人口水平的电子健康记录数据中得出的治疗负担措施相关联1。根据患者的能力进行个性化治疗,患者应该能够更好地坚持护理计划,从而提高治疗期间和治疗后的生活质量。致谢:作者要感谢我们的患者倡导者导师Cheryl Jernigan在这个项目中的指导。我们还要感谢Susan G. Komen基金会对这项研究的支持。引用:1。程,A. C.和Levy, M. A.数据驱动的治疗负担测量方法:乳腺癌患者的研究。AMIA物质。计算机协会。程序。中国生物医学工程学报,2016,37(5):559 - 563。2. Zafar, s.y.等。癌症治疗的经济毒性:一项评估自费费用和投保癌症患者经历的试点研究。肿瘤杂志,18,381-90(2013)。3.梅尔,F. S.和梅,C. R.对治疗负担的思考。[j] .中国医学杂志,2014,(5):444 - 444。4. Boehmer, K. R, Shippee, N. D, Beebe, T. J.和Montori, V. M.追求最小破坏性医学:患者能力与疾病和医疗保健相关需求中断的相关性。j .中国。论文。(2016)。5. Ishii, H.等。测量2型糖尿病患者治疗负担问卷的可重复性和有效性:糖尿病治疗负担问卷(DTBQ)。中国糖尿病杂志,2018,31(5):444 - 444。引用格式:Cheng A, Levy M.乳腺癌患者的治疗负担和护理管理能力[摘要]。2018年圣安东尼奥乳腺癌研讨会论文集;2018年12月4-8日;费城(PA): AACR;癌症杂志,2019;79(4增刊):OT2-10-01。
{"title":"Abstract OT2-10-01: Treatment burden and capacity to manage care among patients with breast cancer","authors":"A. Cheng, M. Levy","doi":"10.1158/1538-7445.SABCS18-OT2-10-01","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT2-10-01","url":null,"abstract":"Patients with breast cancer spend significant time1, effort, and financial resources2 to combat the disease for years after their diagnosis. The large volume of healthcare tasks can cause patients to become overburdened, leading to reduced adherence with care plans and worse outcomes3. On the other hand, certain patient characteristics such as physical resilience, financial well-being, and supportive family environments increase patients9 capacity to manage care4. Assessing treatment burden and capacity when prescribing care has been applied to populations such as diabetes patients5. We are investigating this paradigm in treatment of patients with breast cancer. The goal of this preliminary study is to identify significant factors that contribute to treatment burden, capacity to manage care, and outcomes of overburden for patients with breast cancer. Through literature review, interviews with survivors, and expert panels of navigators and providers, we will develop a survey instrument given to patients at the time of diagnosis. The survey will assess patient capacity and help providers give treatment options based on attributes of the patient. Additionally, we will attempt to correlate survey results with treatment burden measures derived from electronic health record data at a population level1. With treatment personalized for patient capacity, patients should be better able to adhere to care plans leading to improved quality of life during treatment and beyond. Acknowledgements: The authors would like to thank Cheryl Jernigan, our patient advocate mentor, for her guidance in this project. We would also like to thank the Susan G. Komen Foundation for their support of this research. References: 1. Cheng, A. C. & Levy, M. A. Data Driven Approach to Burden of Treatment Measurement: A Study of Patients with Breast Cancer. AMIA Annu. Symp. proceedings. AMIA Symp.2016, 1756–1763 (2016). 2. Zafar, S. Y. et al. The financial toxicity of cancer treatment: a pilot study assessing out-of-pocket expenses and the insured cancer patient9s experience. Oncologist.18, 381–90 (2013). 3. Mair, F. S. & May, C. R. Thinking about the burden of treatment. BMJ.349, g6680–g6680 (2014). 4. Boehmer, K. R., Shippee, N. D., Beebe, T. J. & Montori, V. M. Pursuing Minimally Disruptive Medicine: Correlation of patient capacity with disruption from illness and healthcare-related demands. J. Clin. Epidemiol. (2016). 5. Ishii, H. et al. Reproducibility and Validity of a Questionnaire Measuring Treatment Burden on Patients with Type 2 Diabetes: Diabetic Treatment Burden Questionnaire (DTBQ). Diabetes Ther.9, 1001–1019 (2018). Citation Format: Cheng A, Levy M. Treatment burden and capacity to manage care among patients with breast cancer [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT2-10-01.","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"6 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74603975","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT1-05-02
R. Stein, L. Hughes-Davies, A. Makris, I. Macpherson, C. Conefrey, L. Rooshenas, S. Pinder, J. Thomas, P. Hall, D. Cameron, H. Earl, B. Naume, C. Poole, D. Rea, S. Macintosh, V. Harmer, A. Morgan, C. Hulme, C. McCabe, N. Stallard, Helen B Higgins, J. Donovan, J. Bartlett, A. Marshall, J. Dunn
Background:Multi-parameter tumour gene expression assays (MPAs) are widely used to estimate individual patient residual risk and to guide chemotherapy use in hormone-sensitive, HER2-negative early breast cancer. The TAILORx trial supports MPA use in a node-negative population. Evidence for MPA use in node-positive breast cancer is limited. OPTIMA (Optimal Personalised Treatment of early breast cancer usIng Multi-parameter Analysis) (ISRCTN42400492) aims to validate MPAs as predictors of chemotherapy sensitivity in a largely node-positive breast cancer population where prospective RCT (Randomised Controlled Trial) evidence is lacking. Methods: OPTIMA is a partially blinded multi-center RCT with an adaptive two-stage design. The main eligibility criteria are women and men age 40 or older with resected ER-positive, HER2-negative invasive breast cancer and up to 9 involved axillary lymph nodes. Randomisation is to standard management (chemotherapy and endocrine therapy) or to MPA-directed treatment using the Prosigna (PAM50) test. Those with a Prosigna tumour score (ROR_PT) >60 receive standard management whilst those with a low score (≤60) are treated with endocrine therapy alone. Endocrine therapy for pre-menopausal women includes ovarian suppression. The co-primary outcomes are (1) Invasive Disease Free Survival (IDFS) and (2) cost-effectiveness of test-directed treatment. Secondary outcomes include IDFS in patients with low-score tumours and quality of life. An integrated qualitative recruitment study addresses challenges to consent and recruitment and will build on experience from the feasibility study that a multidisciplinary approach at sites is important for recruitment success. Tumour blocks will be banked to allow evaluation of additional MPA technologies. Recruitment of 4500 patients over 5 years will permit demonstration of 3% non-inferiority of test-directed treatment, assuming 5-year IDFS of 85% with standard management, equivalent to a HR of 1.22. Inclusion of patients from the feasibility study will increase the power to test for non-inferiority. Results: The OPTIMA main trial opened in January 2017. Overall recruitment (including the feasibility study) will reach 1000 in August 2018. Recruitment in Norway will commence in July 2018. Characteristics of the OPTIMA main participants recruited to 31st May 2018 are shown in the table. Conclusion: OPTIMA is one of two large scale prospective trials validating the use of test-guided chemotherapy decisions in node-positive early breast cancer. It is expected to have a global impact on breast cancer treatment. Experience from the preliminary study and close engagement with centres will aid trial success. Funding: OPTIMA is funded by the UK NIHR HTA Programme (10/34/501). Views expressed are those of the authors and not those of the HTA Programme, NIHR, NHS or the DoH. Citation Format: Stein RC, Hughes-Davies L, Makris A, Macpherson IR, Conefrey C, Rooshenas L, Pinder SE, Thomas J, Hall PS, Ca
背景:多参数肿瘤基因表达测定(MPAs)被广泛用于评估个体患者残留风险,并指导激素敏感的her2阴性早期乳腺癌的化疗使用。TAILORx试验支持在淋巴结阴性人群中使用MPA。在淋巴结阳性乳腺癌中使用MPA的证据有限。OPTIMA(使用多参数分析的早期乳腺癌最佳个性化治疗)(ISRCTN42400492)旨在验证MPAs在大部分淋巴结阳性乳腺癌人群中作为化疗敏感性的预测因子,这些人群缺乏前瞻性RCT(随机对照试验)证据。方法:OPTIMA是一项部分盲法多中心随机对照试验,采用自适应两期设计。主要的入选标准是年龄在40岁或以上的女性和男性,并且患有切除的er阳性、her2阴性的浸润性乳腺癌和不超过9个受累的腋窝淋巴结。随机分组为标准管理(化疗和内分泌治疗)或使用Prosigna (PAM50)测试进行mpa指导治疗。Prosigna肿瘤评分(ROR_PT) >60的患者接受标准治疗,评分较低(≤60)的患者单独接受内分泌治疗。绝经前妇女的内分泌治疗包括抑制卵巢。共同主要结局是(1)无侵袭性疾病生存(IDFS)和(2)试验导向治疗的成本效益。次要结局包括低评分肿瘤患者的IDFS和生活质量。一项综合的定性招聘研究解决了同意和招聘方面的挑战,并将根据可行性研究的经验,即在工作地点采取多学科方法对招聘成功很重要。肿瘤块将被储存起来,以便评估额外的MPA技术。在5年内招募4500名患者将证明试验导向治疗的非劣效性为3%,假设采用标准管理的5年IDFS为85%,相当于HR为1.22。纳入可行性研究的患者将增加检验非劣效性的能力。结果:OPTIMA主要试验于2017年1月开始。2018年8月,整体招聘(含可行性研究)将达到1000人。挪威的招聘将于2018年7月开始。截至2018年5月31日,OPTIMA主要参与者的特征见表。结论:OPTIMA是验证在淋巴结阳性早期乳腺癌中使用测试指导化疗决策的两项大规模前瞻性试验之一。预计它将对乳腺癌治疗产生全球性影响。初步研究的经验以及与各中心的密切接触将有助于试验的成功。资金:OPTIMA由英国NIHR HTA项目(10/34/501)资助。本文仅代表作者的观点,不代表HTA项目、NIHR、NHS或DoH的观点。引用格式:Stein RC, Hughes-Davies L, Makris A, Macpherson IR, Conefrey C, Rooshenas L, Pinder SE, Thomas J, Hall PS, Cameron DA, Earl HM, name B, Poole CJ, Rea DW, MacIntosh SA, Harmer V, Morgan A, Hulme C, McCabe C, Stallard N, Higgins H, Donovan JL, Bartlett JM, Marshall A, Dunn JA。OPTIMA:一项前瞻性随机试验,旨在验证基因表达测试指导的化疗决策在临床高风险早期乳腺癌中的临床效用和成本效益。2018年圣安东尼奥乳腺癌研讨会论文集;2018年12月4-8日;费城(PA): AACR;中国癌症杂志,2019;79(4增刊):01-05-02。
{"title":"Abstract OT1-05-02: OPTIMA: A prospective randomized trial to validate the clinical utility and cost-effectiveness of gene expression test-directed chemotherapy decisions in high clinical risk early breast cancer","authors":"R. Stein, L. Hughes-Davies, A. Makris, I. Macpherson, C. Conefrey, L. Rooshenas, S. Pinder, J. Thomas, P. Hall, D. Cameron, H. Earl, B. Naume, C. Poole, D. Rea, S. Macintosh, V. Harmer, A. Morgan, C. Hulme, C. McCabe, N. Stallard, Helen B Higgins, J. Donovan, J. Bartlett, A. Marshall, J. Dunn","doi":"10.1158/1538-7445.SABCS18-OT1-05-02","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT1-05-02","url":null,"abstract":"Background:Multi-parameter tumour gene expression assays (MPAs) are widely used to estimate individual patient residual risk and to guide chemotherapy use in hormone-sensitive, HER2-negative early breast cancer. The TAILORx trial supports MPA use in a node-negative population. Evidence for MPA use in node-positive breast cancer is limited. OPTIMA (Optimal Personalised Treatment of early breast cancer usIng Multi-parameter Analysis) (ISRCTN42400492) aims to validate MPAs as predictors of chemotherapy sensitivity in a largely node-positive breast cancer population where prospective RCT (Randomised Controlled Trial) evidence is lacking. Methods: OPTIMA is a partially blinded multi-center RCT with an adaptive two-stage design. The main eligibility criteria are women and men age 40 or older with resected ER-positive, HER2-negative invasive breast cancer and up to 9 involved axillary lymph nodes. Randomisation is to standard management (chemotherapy and endocrine therapy) or to MPA-directed treatment using the Prosigna (PAM50) test. Those with a Prosigna tumour score (ROR_PT) >60 receive standard management whilst those with a low score (≤60) are treated with endocrine therapy alone. Endocrine therapy for pre-menopausal women includes ovarian suppression. The co-primary outcomes are (1) Invasive Disease Free Survival (IDFS) and (2) cost-effectiveness of test-directed treatment. Secondary outcomes include IDFS in patients with low-score tumours and quality of life. An integrated qualitative recruitment study addresses challenges to consent and recruitment and will build on experience from the feasibility study that a multidisciplinary approach at sites is important for recruitment success. Tumour blocks will be banked to allow evaluation of additional MPA technologies. Recruitment of 4500 patients over 5 years will permit demonstration of 3% non-inferiority of test-directed treatment, assuming 5-year IDFS of 85% with standard management, equivalent to a HR of 1.22. Inclusion of patients from the feasibility study will increase the power to test for non-inferiority. Results: The OPTIMA main trial opened in January 2017. Overall recruitment (including the feasibility study) will reach 1000 in August 2018. Recruitment in Norway will commence in July 2018. Characteristics of the OPTIMA main participants recruited to 31st May 2018 are shown in the table. Conclusion: OPTIMA is one of two large scale prospective trials validating the use of test-guided chemotherapy decisions in node-positive early breast cancer. It is expected to have a global impact on breast cancer treatment. Experience from the preliminary study and close engagement with centres will aid trial success. Funding: OPTIMA is funded by the UK NIHR HTA Programme (10/34/501). Views expressed are those of the authors and not those of the HTA Programme, NIHR, NHS or the DoH. Citation Format: Stein RC, Hughes-Davies L, Makris A, Macpherson IR, Conefrey C, Rooshenas L, Pinder SE, Thomas J, Hall PS, Ca","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"21 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73894206","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT2-06-01
M. Schmidt, S. Bolte, K. Frenzel, L. Heesen, E. Derhovanessian, V. Bukur, M. Diken, J. Gruetzner, S. Kreiter, A. Klein, A. Kuhn, D. Langer, M. Loewer, H. Lindman, A. Schneeweiss, O. Tuereci, U. Şahin
Background: The treatment of triple negative breast cancer (TNBC) is hampered by the lack of established therapeutic targets such as hormone receptors or HER-2. Chemotherapy and radiotherapy is the standard of care, yet survival rates in TNBC remain poor. Approaches tailored to the patient9s individual tumor signature may lead to improved therapeutic outcome. We have set up a clinical workflow covering drug development (from target discovery to manufacturing) and drug release providing a custom-made investigational medicinal product (IMP) for each individual patient. Trial Design: A phase I/II trial assesses the feasibility, safety and biological efficacy of this personalized immunotherapy in three clinical sites in Germany and Sweden. TNBC patients (pT1cN0M0 – TxNxM0) after completion of initial standard of care therapy will be allocated to one of two study arms. Patients in ARM1 receive 8 vaccination cycles with a personalized combination of shared tumor-associated antigens, selected based on each patient tumor9s antigen-expression profile out of a WAREHOUSE of pre-manufactured mRNA vaccine. Patients in ARM2 receive the personalized mRNA WAREHOUSE vaccine followed by 8 vaccination cycles of an on-demand manufactured mRNA MUTANOME vaccine encoding up to twenty unique neo-epitopes of the individual patient identified by next generation sequencing. The mRNAs are administered intravenously as a nanoparticulate lipoplex formulation, which protects RNA from degradation, activates innate immunity, transfects APCs and consequently induces highly potent antigen-specific T-cell responses. The treatment of 12 patients in ARM1 is completed and enrolment of patients for ARM2 has started. Preliminary data show that the RNA-WAREHOUSE approach is feasible and can be applied safely. Biomarker analysis is ongoing. This approach is promising as it addresses the heterogeneity of TNBC. The TNBC-MERIT trial was initially funded by the EU Commission9s FP7 and led by BioNTech AG. Citation Format: Schmidt M, Bolte S, Frenzel K, Heesen L, Derhovanessian E, Bukur V, Diken M, Gruetzner J, Kreiter S, Klein A, Kuhn A, Langer D, Loewer M, Lindman H, Schneeweiss A, Tuereci O, Sahin U. Highly innovative personalized RNA-immunotherapy for patients with triple negative breast cancer [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT2-06-01.
背景:三阴性乳腺癌(TNBC)的治疗受到缺乏既定治疗靶点(如激素受体或HER-2)的阻碍。化疗和放疗是标准的治疗方法,但TNBC的生存率仍然很低。针对患者个体肿瘤特征量身定制的方法可能会改善治疗效果。我们已经建立了涵盖药物开发(从目标发现到生产)和药物释放的临床工作流程,为每位患者提供定制的研究药物(IMP)。试验设计:一项I/II期试验在德国和瑞典的三个临床地点评估了这种个性化免疫疗法的可行性、安全性和生物学有效性。TNBC患者(pT1cN0M0 - TxNxM0)在完成初始标准护理治疗后将被分配到两个研究组中的一个。ARM1患者接受8个接种周期,使用共享肿瘤相关抗原的个性化组合,这些抗原是根据每个患者的肿瘤抗原表达谱从预先制造的mRNA疫苗中选择的。ARM2患者接受个体化mRNA WAREHOUSE疫苗,随后接受8次按需制造的mRNA MUTANOME疫苗接种周期,该疫苗编码多达20个由下一代测序确定的个体患者的独特新表位。这些mrna以纳米粒脂质体的形式静脉注射,可以保护RNA免受降解,激活先天免疫,转染apc,从而诱导高效的抗原特异性t细胞反应。12例ARM1患者的治疗已经完成,ARM2患者的登记工作已经开始。初步数据表明,RNA-WAREHOUSE方法是可行的,可以安全应用。生物标志物分析正在进行中。这种方法很有希望,因为它解决了TNBC的异质性。TNBC-MERIT试验最初由欧盟委员会FP7资助,由BioNTech AG公司领导。引用格式:Schmidt M, Bolte S, Frenzel K, Heesen L, Derhovanessian E, Bukur V, Diken M, Gruetzner J, Kreiter S, Klein A, Kuhn A, Langer D, Loewer M, Lindman H, Schneeweiss A, Tuereci O, Sahin u。三阴性乳腺癌患者的个性化rna免疫治疗[摘要]。2018年圣安东尼奥乳腺癌研讨会论文集;2018年12月4-8日;费城(PA): AACR;中国癌症杂志2019;79(4增刊):OT2-06-01。
{"title":"Abstract OT2-06-01: Highly innovative personalized RNA-immunotherapy for patients with triple negative breast cancer","authors":"M. Schmidt, S. Bolte, K. Frenzel, L. Heesen, E. Derhovanessian, V. Bukur, M. Diken, J. Gruetzner, S. Kreiter, A. Klein, A. Kuhn, D. Langer, M. Loewer, H. Lindman, A. Schneeweiss, O. Tuereci, U. Şahin","doi":"10.1158/1538-7445.SABCS18-OT2-06-01","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT2-06-01","url":null,"abstract":"Background: The treatment of triple negative breast cancer (TNBC) is hampered by the lack of established therapeutic targets such as hormone receptors or HER-2. Chemotherapy and radiotherapy is the standard of care, yet survival rates in TNBC remain poor. Approaches tailored to the patient9s individual tumor signature may lead to improved therapeutic outcome. We have set up a clinical workflow covering drug development (from target discovery to manufacturing) and drug release providing a custom-made investigational medicinal product (IMP) for each individual patient. Trial Design: A phase I/II trial assesses the feasibility, safety and biological efficacy of this personalized immunotherapy in three clinical sites in Germany and Sweden. TNBC patients (pT1cN0M0 – TxNxM0) after completion of initial standard of care therapy will be allocated to one of two study arms. Patients in ARM1 receive 8 vaccination cycles with a personalized combination of shared tumor-associated antigens, selected based on each patient tumor9s antigen-expression profile out of a WAREHOUSE of pre-manufactured mRNA vaccine. Patients in ARM2 receive the personalized mRNA WAREHOUSE vaccine followed by 8 vaccination cycles of an on-demand manufactured mRNA MUTANOME vaccine encoding up to twenty unique neo-epitopes of the individual patient identified by next generation sequencing. The mRNAs are administered intravenously as a nanoparticulate lipoplex formulation, which protects RNA from degradation, activates innate immunity, transfects APCs and consequently induces highly potent antigen-specific T-cell responses. The treatment of 12 patients in ARM1 is completed and enrolment of patients for ARM2 has started. Preliminary data show that the RNA-WAREHOUSE approach is feasible and can be applied safely. Biomarker analysis is ongoing. This approach is promising as it addresses the heterogeneity of TNBC. The TNBC-MERIT trial was initially funded by the EU Commission9s FP7 and led by BioNTech AG. Citation Format: Schmidt M, Bolte S, Frenzel K, Heesen L, Derhovanessian E, Bukur V, Diken M, Gruetzner J, Kreiter S, Klein A, Kuhn A, Langer D, Loewer M, Lindman H, Schneeweiss A, Tuereci O, Sahin U. Highly innovative personalized RNA-immunotherapy for patients with triple negative breast cancer [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT2-06-01.","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"27 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82844374","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT1-11-01
J. Huober, G. Nagel, A. Rempen, E. Schlicht, F. Flock, S. Fritz, F. Thiel, L. Wiesmüller, R. Felderbaum, V. Heilmann, I. Bekes, V. Fink, S. Albrecht, N. Gregorio, M. Tzschaschel, K. Ernst, C. Wolf, P. Kuhn, T. Friedl, W. Janni, A. D. Gregorio
Background: Further progress in the treatment of breast cancer will likely come from contributions of molecular biology and immunologic approaches. The search for druggable molecular aberrations may enable treatment based on the molecular profile. A better identification of patients with a high risk of relapse facilitates the selection of these pts for clinical trials investigating early therapeutic molecular-based interventions. Trial Design: The BRandO BiO Registry is a multi-center regional registry to record clinical, epidemiological, and biological data from patients with newly diagnosed breast and ovarian cancer at the University of Ulm, Dept. of Gynecology and 19 affiliated network hospitals and practices in the Alb-Allgau Bodensee region (outreach area of the Comprehensive Cancer Center Ulm). Longitudinal biobanking is included with collection of paraffin-embedded samples of the primary tumor as well as blood samples at first diagnosis, after 6 and 12 months and at first relapse to isolate and investigate cell-free and germline DNA. Epidemiological, life style and quality of life (QOL) questionnaires are collected at first diagnosis, after 12, 36 and 60 months. The follow up is planned for 10 years. Eligibility criteria: Patients with primary newly diagnosed untreated breast or ovarian cancer of ≥ 18 years are eligible; primary metastatic untreated disease is allowed. Exclusion criteria comprise severe neurological or psychiatric disorders interfering with the ability to give an informed consent, no consent for registration, storage and processing of the individual disease characteristics and bio samples, and any malignant tumor in the last 3 years (except in situ disease). Specific aims: To register the majority of patients with newly diagnosed breast or ovarian cancer in all BRandO-BiO participating centers of a well-defined geographical area. To assess clinical characteristics and outcome data (event-free survival, overall survival) of these patients. To evaluate the primary tumor of all patients for mutational (druggable) aberrations. Further to assess cell-free DNA in the serial blood samples at baseline, 6 and 12 months and correlate these results with clinical outcome data as well as tumor and patient characteristics to look for early markers predicting relapse. To perform a longitudinal assessment of the patients9 sociodemographic factors, comorbidities, lifestyle and QOL factors by analyzing serial questionnaires collected at recruitment and at 12, 36 and 60 months. Present accrual and target accrual: The BRandO BiO Registry started January 2016 in the Dept. of Gynecology, University of Ulm and February 2017 at the network hospitals and practices. Until June 2018, 1180 patients with primary breast or ovarian cancer have been enrolled. The current adherence to serial blood testing and serial questionnaires is good with a return rate of 90%. A sample size of 3000 patients is planned. Contact information: Jens Huober, University of
背景:乳腺癌治疗的进一步进展可能来自分子生物学和免疫学方法的贡献。对可药物分子畸变的研究可能使基于分子谱的治疗成为可能。更好地识别复发风险高的患者有助于选择这些患者进行临床试验,研究早期治疗性分子干预措施。试验设计:BRandO生物登记处是一个多中心区域登记处,记录乌尔姆大学妇科和19家附属网络医院新诊断的乳腺癌和卵巢癌患者的临床、流行病学和生物学数据,并在Alb-Allgau Bodensee地区(乌尔姆综合癌症中心的外展区域)进行实践。纵向生物银行包括收集原发肿瘤的石蜡包埋样本以及首次诊断时、6个月和12个月后以及首次复发时的血液样本,以分离和研究无细胞和种系DNA。首次诊断时、12个月、36个月和60个月分别收集流行病学、生活方式和生活质量(QOL)问卷。后续研究计划进行10年。入选标准:原发新诊断未经治疗的乳腺癌或卵巢癌患者≥18年;原发转移性未治疗疾病是允许的。排除标准包括严重的神经或精神疾病,干扰了知情同意的能力,不同意个人疾病特征和生物样本的注册、存储和处理,以及过去3年内任何恶性肿瘤(原位疾病除外)。具体目的:在明确地理区域的所有BRandO-BiO参与中心登记大多数新诊断的乳腺癌或卵巢癌患者。评估这些患者的临床特征和结局数据(无事件生存期、总生存期)。评估所有患者原发肿瘤的突变(可用药)异常。进一步评估基线、6个月和12个月系列血液样本中的无细胞DNA,并将这些结果与临床结果数据以及肿瘤和患者特征相关联,以寻找预测复发的早期标志物。通过分析招募时、12个月、36个月和60个月收集的系列问卷,对患者的9个社会人口因素、合并症、生活方式和生活质量因素进行纵向评估。当前收益和目标收益:BRandO生物注册于2016年1月在乌尔姆大学妇科开始,2017年2月在网络医院和实践中开始。截至2018年6月,已有1180名原发性乳腺癌或卵巢癌患者入组。目前对系列血液检测和系列问卷的依从性良好,回收率为90%。计划的样本量为3000名患者。联系方式:Jens Huober,乌尔姆大学,妇科,乳房中心,jens.huober@uniklinik-ulm.de Amelie de Gregorio,乌尔姆大学,妇科,乳房中心,Amelie.de Gregorio@uniklinik-ulm.deHuober J, Nagel G, Rempen A, Schlicht E, Flock F, Fritz S, Thiel F, Wiesmuller L, Felderbaum R, Heilmann V, Bekes I, Fink V, Albrecht S, De Gregorio N, Tzschaschel M, Ernst K, Wolf C, Kuhn P, Friedl T, Janni W, De Gregorio A.基于生物库的原发性乳腺癌和卵巢癌患者流行病学、生活方式和生活质量因素评价的多中心区域登记[摘要]。2018年圣安东尼奥乳腺癌研讨会论文集;2018年12月4-8日;费城(PA): AACR;中国癌症杂志,2019;79(4增刊):1-11-01。
{"title":"Abstract OT1-11-01: The BRandO BiO registry – A multicenter regional registry for patients with primary breast and ovarian cancer with longitudinal biobanking and evaluation of epidemiological, life style and quality of life factors","authors":"J. Huober, G. Nagel, A. Rempen, E. Schlicht, F. Flock, S. Fritz, F. Thiel, L. Wiesmüller, R. Felderbaum, V. Heilmann, I. Bekes, V. Fink, S. Albrecht, N. Gregorio, M. Tzschaschel, K. Ernst, C. Wolf, P. Kuhn, T. Friedl, W. Janni, A. D. Gregorio","doi":"10.1158/1538-7445.SABCS18-OT1-11-01","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT1-11-01","url":null,"abstract":"Background: Further progress in the treatment of breast cancer will likely come from contributions of molecular biology and immunologic approaches. The search for druggable molecular aberrations may enable treatment based on the molecular profile. A better identification of patients with a high risk of relapse facilitates the selection of these pts for clinical trials investigating early therapeutic molecular-based interventions. Trial Design: The BRandO BiO Registry is a multi-center regional registry to record clinical, epidemiological, and biological data from patients with newly diagnosed breast and ovarian cancer at the University of Ulm, Dept. of Gynecology and 19 affiliated network hospitals and practices in the Alb-Allgau Bodensee region (outreach area of the Comprehensive Cancer Center Ulm). Longitudinal biobanking is included with collection of paraffin-embedded samples of the primary tumor as well as blood samples at first diagnosis, after 6 and 12 months and at first relapse to isolate and investigate cell-free and germline DNA. Epidemiological, life style and quality of life (QOL) questionnaires are collected at first diagnosis, after 12, 36 and 60 months. The follow up is planned for 10 years. Eligibility criteria: Patients with primary newly diagnosed untreated breast or ovarian cancer of ≥ 18 years are eligible; primary metastatic untreated disease is allowed. Exclusion criteria comprise severe neurological or psychiatric disorders interfering with the ability to give an informed consent, no consent for registration, storage and processing of the individual disease characteristics and bio samples, and any malignant tumor in the last 3 years (except in situ disease). Specific aims: To register the majority of patients with newly diagnosed breast or ovarian cancer in all BRandO-BiO participating centers of a well-defined geographical area. To assess clinical characteristics and outcome data (event-free survival, overall survival) of these patients. To evaluate the primary tumor of all patients for mutational (druggable) aberrations. Further to assess cell-free DNA in the serial blood samples at baseline, 6 and 12 months and correlate these results with clinical outcome data as well as tumor and patient characteristics to look for early markers predicting relapse. To perform a longitudinal assessment of the patients9 sociodemographic factors, comorbidities, lifestyle and QOL factors by analyzing serial questionnaires collected at recruitment and at 12, 36 and 60 months. Present accrual and target accrual: The BRandO BiO Registry started January 2016 in the Dept. of Gynecology, University of Ulm and February 2017 at the network hospitals and practices. Until June 2018, 1180 patients with primary breast or ovarian cancer have been enrolled. The current adherence to serial blood testing and serial questionnaires is good with a return rate of 90%. A sample size of 3000 patients is planned. Contact information: Jens Huober, University of ","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"30 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"81382400","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT3-06-02
M. Radovich, Jeffrey P. Solzak, B. Hancock, A. Storniolo, B. Schneider, K. Miller
Background: The PI3K pathway is dysregulated in the majority of triple-negative breast cancer (TNBCs). Contrary to the theory of oncogene addiction, single agent inhibition of the PI3K pathway in TNBC has had only modest activity. Our group has demonstrated preclinically that when PI3K is inhibited, an immediate compensatory up-regulation of the Wnt pathway occurs. The Wnt pathway is known known for its role in cancer metastases and can confer resistance to initial PI3K inhibition. Simultaneous dual targeting of both pathways is highly synergistic against TNBC models in vitro and in vivo. We have initiated a Phase I clinical trial using Gedatolisib (PI3K/mTOR inhibitor) and PTK7-ADC (Wnt pathway) for patients with metastatic TNBC (NCT03243331). Gedatolisib is a pan-class I isoform PI3K/mTOR inhibitor, and PTK7-ADC is an antibody-drug conjugate against the cell-surface PTK7 protein (Wnt pathway co-receptor) with an Auristatin payload. PTK7 is an attractive second target due to its up-regulation after PI3K inhibition and its known overexpression in TNBC. Further data has shown that the PTK7-payload, Auristatin, is in itself synergistic with Gedatolisib. The combination of using both of these drugs suggests a unique concept of “double synergy”. Where Gedatolisib increases the expression of the target of PTK7-ADC leading to one mechanism of synergy, and the Auristatin payload on PTK7-ADC is synergistic with Gedatolisib providing a second mechanism. Study Design: This is an open-label, Phase I, dose-escalation study with a 3 + 3 cohort design. The trial will enroll 12-18 patients. 3 cohorts of at least 3 patients will receive Gedatolisib (weekly) & PTK-ADC (q3w) at 110mg+1.4mg/kg, 180mg+1.4mg/kg, and 180mg+2.8mg/kg dose levels. Eligibility Criteria: This trial enrolls patients with metastatic triple negative (ER-, PgR-, HER2-) or low estrogen expressing (ER and PgR Objectives: The primary objective is to evaluate the safety of Gedatolisib plus PTK7-ADC. The secondary objective is to evaluate efficacy as determined by objective response rate, clinical benefit at 18 weeks, and progression free survival (PFS). Exploratory objectives will evaluate efficacy in patients with genomic aberrations in the PI3K pathway; and association of tumor DNA, RNA, plasma and circulating tumor cell sequencing with clinical efficacy to identify putative biomarkers. Correlative Sciences: We are collecting matched pre-/post-treatment tumor biopsies and serial blood samples to determine biomarkers of clinical response to inform subsequent trials. We plan to evaluate: 1) PI3K activity; 2) genomic aberrations in the PI3K pathway; 3) baseline PTK7 expression; 4) PTK7 upregulation after Gedatolisib treatment; and 5) mutations in plasma circulating tumor DNA. Supported by the BCRF, 100 Voices of Hope, Catherine Peachey Foundation, and Pfizer. Citation Format: Radovich M, Solzak JP, Hancock BA, Storniolo AMV, Schneider BP, Miller KD. An initial safety study of gedatolisib plus PTK7-
{"title":"Abstract OT3-06-02: An initial safety study of gedatolisib plus PTK7-ADC for metastatic triple-negative breast cancer","authors":"M. Radovich, Jeffrey P. Solzak, B. Hancock, A. Storniolo, B. Schneider, K. Miller","doi":"10.1158/1538-7445.SABCS18-OT3-06-02","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT3-06-02","url":null,"abstract":"Background: The PI3K pathway is dysregulated in the majority of triple-negative breast cancer (TNBCs). Contrary to the theory of oncogene addiction, single agent inhibition of the PI3K pathway in TNBC has had only modest activity. Our group has demonstrated preclinically that when PI3K is inhibited, an immediate compensatory up-regulation of the Wnt pathway occurs. The Wnt pathway is known known for its role in cancer metastases and can confer resistance to initial PI3K inhibition. Simultaneous dual targeting of both pathways is highly synergistic against TNBC models in vitro and in vivo. We have initiated a Phase I clinical trial using Gedatolisib (PI3K/mTOR inhibitor) and PTK7-ADC (Wnt pathway) for patients with metastatic TNBC (NCT03243331). Gedatolisib is a pan-class I isoform PI3K/mTOR inhibitor, and PTK7-ADC is an antibody-drug conjugate against the cell-surface PTK7 protein (Wnt pathway co-receptor) with an Auristatin payload. PTK7 is an attractive second target due to its up-regulation after PI3K inhibition and its known overexpression in TNBC. Further data has shown that the PTK7-payload, Auristatin, is in itself synergistic with Gedatolisib. The combination of using both of these drugs suggests a unique concept of “double synergy”. Where Gedatolisib increases the expression of the target of PTK7-ADC leading to one mechanism of synergy, and the Auristatin payload on PTK7-ADC is synergistic with Gedatolisib providing a second mechanism. Study Design: This is an open-label, Phase I, dose-escalation study with a 3 + 3 cohort design. The trial will enroll 12-18 patients. 3 cohorts of at least 3 patients will receive Gedatolisib (weekly) & PTK-ADC (q3w) at 110mg+1.4mg/kg, 180mg+1.4mg/kg, and 180mg+2.8mg/kg dose levels. Eligibility Criteria: This trial enrolls patients with metastatic triple negative (ER-, PgR-, HER2-) or low estrogen expressing (ER and PgR Objectives: The primary objective is to evaluate the safety of Gedatolisib plus PTK7-ADC. The secondary objective is to evaluate efficacy as determined by objective response rate, clinical benefit at 18 weeks, and progression free survival (PFS). Exploratory objectives will evaluate efficacy in patients with genomic aberrations in the PI3K pathway; and association of tumor DNA, RNA, plasma and circulating tumor cell sequencing with clinical efficacy to identify putative biomarkers. Correlative Sciences: We are collecting matched pre-/post-treatment tumor biopsies and serial blood samples to determine biomarkers of clinical response to inform subsequent trials. We plan to evaluate: 1) PI3K activity; 2) genomic aberrations in the PI3K pathway; 3) baseline PTK7 expression; 4) PTK7 upregulation after Gedatolisib treatment; and 5) mutations in plasma circulating tumor DNA. Supported by the BCRF, 100 Voices of Hope, Catherine Peachey Foundation, and Pfizer. Citation Format: Radovich M, Solzak JP, Hancock BA, Storniolo AMV, Schneider BP, Miller KD. An initial safety study of gedatolisib plus PTK7-","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"29 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83574117","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT2-01-01
B. Yang, Ju-fang Wu
This abstract was not presented at the conference. Citation Format: Yang B, Wu J. Not presented [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT2-01-01.
{"title":"Abstract OT2-01-01: Not presented","authors":"B. Yang, Ju-fang Wu","doi":"10.1158/1538-7445.SABCS18-OT2-01-01","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT2-01-01","url":null,"abstract":"This abstract was not presented at the conference. Citation Format: Yang B, Wu J. Not presented [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT2-01-01.","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"2014 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88042443","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT1-01-02
P. Plourde, L. Schwartzberg, G. Greene, D. Portman, B. Komm, Sn Jenkins, P. Liu, Portman, M. Goetz
Endocrine based therapy is the standard treatment for estrogen receptor positive (ER+) MBC. Agents targeting the ER pathway including aromatase inhibitors (AIs), fulvestrant and tamoxifen along with CDK 4/6 inhibitors are considered standard for first and 2nd line treatment. However, endocrine resistance develops in nearly all patients and the optimal systemic therapy after progression on a CDK 4/6 inhibitor is unknown. Lasofoxifene is a third generation SERM previously investigated for the treatment of osteoporosis and vulvo-vaginal atrophy (VVA). In a large phase 3 trial evaluating the efficacy of lasofoxifene for the postmenopausal treatment of osteoporosis, lasofoxifene significantly reduced the incidence of ER+ breast cancer. Further unpublished preclinical data have demonstrated significant in vitro and in vivo efficacy in non-clinical breast cancer models including models with and without ESR1 mutants. Moreover, lasofoxifene significantly reduced metastases in ESR1 mutated models. These non-clinical and clinical data provide a strong rationale to pursue a phase 2 clinical trial in women with ER+, ESR1 mutated MBC. This open-label, multi-center study will compare the efficacy and tolerability of lasofoxifene (5 mg orally daily) to fulvestrant (IM 500 mg D1,15,29 and then q30 D) in a 1:1 randomization. Inclusion criteria include postmenopausal women with ER+ advanced breast cancer; progression on a non-steroidal AI in combination with a CDK 4/6 inhibitor; and a known ESR1 mutation. Approximately 90 patients with measurable or evaluable disease (i.e. bone only) will be recruited to have at least 40 patients per treatment arm. The primary endpoint will be progression free survival (PFS) with secondary endpoints of objective response rate (ORR), clinical benefit rate (CBR), duration of response (DoR) and time to response (TTR). It is assumed that lasofoxifene will double the median PFS compared to fulvestrant in this ESR1 mutation patient population for a hazard ratio 0.5 and a power of 89% to reach a 1-sided p of The study will commence in 4Q2018 and will complete recruitment in 1 year. It is anticipated that 25-30 centers in the US will be participating. Citation Format: Plourde PV, Schwartzberg LS, Greene GL, Portman DJ, Komm BS, Jenkins SN, Liu P-Y, Portman MD, Goetz MP. An open-label, randomized, multi-center phase 2 study evaluating the activity of lasofoxifene relative to fulvestrant for the treatment of postmenopausal women with locally advanced or metastatic ER+/HER2 - breast cancer (MBC) with an ESR1 mutation [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT1-01-02.
{"title":"Abstract OT1-01-02: An open-label, randomized, multi-center phase 2 study evaluating the activity of lasofoxifene relative to fulvestrant for the treatment of postmenopausal women with locally advanced or metastatic ER+/HER2 - breast cancer (MBC) with an ESR1 mutation","authors":"P. Plourde, L. Schwartzberg, G. Greene, D. Portman, B. Komm, Sn Jenkins, P. Liu, Portman, M. Goetz","doi":"10.1158/1538-7445.SABCS18-OT1-01-02","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT1-01-02","url":null,"abstract":"Endocrine based therapy is the standard treatment for estrogen receptor positive (ER+) MBC. Agents targeting the ER pathway including aromatase inhibitors (AIs), fulvestrant and tamoxifen along with CDK 4/6 inhibitors are considered standard for first and 2nd line treatment. However, endocrine resistance develops in nearly all patients and the optimal systemic therapy after progression on a CDK 4/6 inhibitor is unknown. Lasofoxifene is a third generation SERM previously investigated for the treatment of osteoporosis and vulvo-vaginal atrophy (VVA). In a large phase 3 trial evaluating the efficacy of lasofoxifene for the postmenopausal treatment of osteoporosis, lasofoxifene significantly reduced the incidence of ER+ breast cancer. Further unpublished preclinical data have demonstrated significant in vitro and in vivo efficacy in non-clinical breast cancer models including models with and without ESR1 mutants. Moreover, lasofoxifene significantly reduced metastases in ESR1 mutated models. These non-clinical and clinical data provide a strong rationale to pursue a phase 2 clinical trial in women with ER+, ESR1 mutated MBC. This open-label, multi-center study will compare the efficacy and tolerability of lasofoxifene (5 mg orally daily) to fulvestrant (IM 500 mg D1,15,29 and then q30 D) in a 1:1 randomization. Inclusion criteria include postmenopausal women with ER+ advanced breast cancer; progression on a non-steroidal AI in combination with a CDK 4/6 inhibitor; and a known ESR1 mutation. Approximately 90 patients with measurable or evaluable disease (i.e. bone only) will be recruited to have at least 40 patients per treatment arm. The primary endpoint will be progression free survival (PFS) with secondary endpoints of objective response rate (ORR), clinical benefit rate (CBR), duration of response (DoR) and time to response (TTR). It is assumed that lasofoxifene will double the median PFS compared to fulvestrant in this ESR1 mutation patient population for a hazard ratio 0.5 and a power of 89% to reach a 1-sided p of The study will commence in 4Q2018 and will complete recruitment in 1 year. It is anticipated that 25-30 centers in the US will be participating. Citation Format: Plourde PV, Schwartzberg LS, Greene GL, Portman DJ, Komm BS, Jenkins SN, Liu P-Y, Portman MD, Goetz MP. An open-label, randomized, multi-center phase 2 study evaluating the activity of lasofoxifene relative to fulvestrant for the treatment of postmenopausal women with locally advanced or metastatic ER+/HER2 - breast cancer (MBC) with an ESR1 mutation [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT1-01-02.","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"1 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"81992296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT2-09-01
Trivedi, G. Samimi, J. Wright, K. Holcomb, J. Garber, N. Horowitz, N. Arber, E. Friedman, R. Wenham, Margaret House, H. Parnes, J. Lee, S. Abutaseh, L. Vornik, B. Heckman-Stoddard, P. Brown, K. Crew
Background: Denosumab is a monoclonal antibody that inhibits RANKL and is approved for the prevention of fractures in patients with osteoporosis or bone metastases. The RANKL signaling pathway is also involved in BRCA1-associated mammary tumorigenesis via a progesterone-induced paracrine effect of RANKL on luminal progenitor cells. Pre-clinical studies have demonstrated that RANKL inhibition resulted in reduced proliferation of mammary tumors. Early findings from an ongoing pre-surgical study demonstrated that denosumab treatment resulted in decreased Ki67 proliferation index in benign breast tissue. Based on these data, denosumab is being pursued as a potential preventive agent for breast cancer in BRCA1 mutation carriers. While promising, the effect of RANKL inhibition on gynecologic tissues such as the ovaries and fallopian tubes, in which progesterone has a protective effect, is unknown. Trial design: We will conduct a multicenter, open-label randomized pilot study of presurgical administration of denosumab versus no treatment in premenopausal women with BRCA1/2 mutations undergoing risk-reducing salpingo-oophorectomy (RRSO). A total of 60 women will be randomized 1:1 to Arm 1) 3-4 doses of 120 mg denosumab subcutaneously every 4 weeks or Arm 2) No treatment. Participants will be stratified by 1) BRCA1 versus BRCA2 mutation status and 2) Use of hormonal contraceptives within the past 3 months (yes/no). Assuming a 10% unevaluable rate, we expect to have 54 evaluable participants (27 per arm). Eligibility criteria: 1) Premenopausal women (defined as 18 years; 2) Documented germline pathogenic mutation or likely pathogenic variant in the BRCA1 or BRCA2 gene; 3) Plan for RRSO with or without hysterectomy; 4) ECOG performance status ≤ 1 (Karnofsky ≥ 70%); 5) Normal organ and marrow function; 6) Negative pregnancy test and use of adequate contraception; 7) Willingness to take supplemental oral calcium and vitamin D3; 8) Dental examination within 6 months of enrollment and no evidence of active dental issues; 9) Ability to understand and willingness to provide informed consent. Specific aims: Our primary objective is to compare the effect of denosumab to no treatment on Ki67 expression in the fimbrial end of the fallopian tube. Secondary objectives are to assess Ki67 in ovary and endometrium; cleaved caspase-3, RANK/RANKL, ER/PR, CD44, and STAT3/pSTAT3 expression in fallopian tube, ovary, and endometrium; gene expression profiling in the fallopian tube and ovary; serum markers (progesterone, estradiol, C-terminal telopeptide) and denosumab levels; and toxicity. Statistical methods: The primary endpoint is post-treatment Ki67 expression in the fimbrial end of the fallopian tube in the denosumab arm compared to the no treatment arm. Assuming a standard deviation of 5.0%, we will have 82% power to detect a 4.0% absolute difference (or effect size of 0.8) in Ki67 proliferation index between the denosumab and no treatment groups by applying a 2-sample t-
背景:Denosumab是一种抑制RANKL的单克隆抗体,被批准用于骨质疏松症或骨转移患者的骨折预防。RANKL信号通路还通过黄体酮诱导的RANKL对腔内祖细胞的旁分泌作用参与brca1相关乳腺肿瘤的发生。临床前研究表明,抑制RANKL可减少乳腺肿瘤的增殖。一项正在进行的术前研究的早期发现表明,denosumab治疗可降低良性乳腺组织中的Ki67增殖指数。基于这些数据,denosumab正在作为BRCA1突变携带者乳腺癌的潜在预防药物进行研究。RANKL抑制对卵巢和输卵管等妇科组织的影响虽然有希望,但黄体酮在这些组织中具有保护作用,目前尚不清楚。试验设计:我们将开展一项多中心、开放标签的随机试验研究,对接受降低风险输卵管卵巢切除术(RRSO)的BRCA1/2突变的绝经前妇女进行术前给药denosumab与不给药的对比。共有60名妇女将按1:1的比例随机分配到第1组,每4周皮下注射3-4次120mg地诺单抗;第2组,不治疗。参与者将根据1)BRCA1与BRCA2突变状态和2)过去3个月内使用激素避孕药(是/否)进行分层。假设10%的不可评估率,我们预计有54名可评估的参与者(每组27名)。1)绝经前妇女(定义为18岁;2) BRCA1或BRCA2基因中记录的种系致病性突变或可能的致病性变异;3)子宫切除或不切除的RRSO计划;4) ECOG性能状态≤1 (Karnofsky≥70%);5)器官、骨髓功能正常;6)妊娠试验阴性并采取适当避孕措施;7)愿意补充口服钙和维生素D3;8)入学后6个月内进行牙科检查,无牙齿问题;9)能够理解并愿意提供知情同意。具体目的:我们的主要目的是比较denosumab治疗和不治疗对输卵管缘端Ki67表达的影响。次要目的是评估卵巢和子宫内膜的Ki67;cleaved caspase-3、RANK/RANKL、ER/PR、CD44和STAT3/pSTAT3在输卵管、卵巢和子宫内膜中的表达;输卵管和卵巢基因表达谱的研究血清标志物(孕酮、雌二醇、c端端肽)和地诺单抗水平;和毒性。统计学方法:主要终点是denosumab组与未治疗组相比,治疗后输卵管边缘Ki67的表达。假设标准差为5.0%,我们将有82%的能力通过应用0.05显著性水平的双样本t检验来检测denosumab组和无治疗组之间Ki67增殖指数的4.0%绝对差异(或效应大小为0.8)。目标收益:60名参与者,将于2018年夏季启动。引用格式:Trivedi MS, Samimi G, Wright JD, Holcomb K, Garber JE, Horowitz NS, Arber N, Friedman E, Wenham RM, House M, Parnes H, Lee JJ, Abutaseh S, Vornik LA, Heckman-Stoddard BM, Brown PH, Crew KD。地诺单抗在BRCA1/2突变携带者低风险输卵管卵巢切除术中的初步研究[摘要]。2018年圣安东尼奥乳腺癌研讨会论文集;2018年12月4-8日;费城(PA): AACR;中国癌症杂志,2019;79(4增刊):OT2-09-01。
{"title":"Abstract OT2-09-01: Pilot study of denosumab inBRCA1/2mutation carriers scheduling for risk-reducing salpingo-oophorectomy","authors":"Trivedi, G. Samimi, J. Wright, K. Holcomb, J. Garber, N. Horowitz, N. Arber, E. Friedman, R. Wenham, Margaret House, H. Parnes, J. Lee, S. Abutaseh, L. Vornik, B. Heckman-Stoddard, P. Brown, K. Crew","doi":"10.1158/1538-7445.SABCS18-OT2-09-01","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT2-09-01","url":null,"abstract":"Background: Denosumab is a monoclonal antibody that inhibits RANKL and is approved for the prevention of fractures in patients with osteoporosis or bone metastases. The RANKL signaling pathway is also involved in BRCA1-associated mammary tumorigenesis via a progesterone-induced paracrine effect of RANKL on luminal progenitor cells. Pre-clinical studies have demonstrated that RANKL inhibition resulted in reduced proliferation of mammary tumors. Early findings from an ongoing pre-surgical study demonstrated that denosumab treatment resulted in decreased Ki67 proliferation index in benign breast tissue. Based on these data, denosumab is being pursued as a potential preventive agent for breast cancer in BRCA1 mutation carriers. While promising, the effect of RANKL inhibition on gynecologic tissues such as the ovaries and fallopian tubes, in which progesterone has a protective effect, is unknown. Trial design: We will conduct a multicenter, open-label randomized pilot study of presurgical administration of denosumab versus no treatment in premenopausal women with BRCA1/2 mutations undergoing risk-reducing salpingo-oophorectomy (RRSO). A total of 60 women will be randomized 1:1 to Arm 1) 3-4 doses of 120 mg denosumab subcutaneously every 4 weeks or Arm 2) No treatment. Participants will be stratified by 1) BRCA1 versus BRCA2 mutation status and 2) Use of hormonal contraceptives within the past 3 months (yes/no). Assuming a 10% unevaluable rate, we expect to have 54 evaluable participants (27 per arm). Eligibility criteria: 1) Premenopausal women (defined as 18 years; 2) Documented germline pathogenic mutation or likely pathogenic variant in the BRCA1 or BRCA2 gene; 3) Plan for RRSO with or without hysterectomy; 4) ECOG performance status ≤ 1 (Karnofsky ≥ 70%); 5) Normal organ and marrow function; 6) Negative pregnancy test and use of adequate contraception; 7) Willingness to take supplemental oral calcium and vitamin D3; 8) Dental examination within 6 months of enrollment and no evidence of active dental issues; 9) Ability to understand and willingness to provide informed consent. Specific aims: Our primary objective is to compare the effect of denosumab to no treatment on Ki67 expression in the fimbrial end of the fallopian tube. Secondary objectives are to assess Ki67 in ovary and endometrium; cleaved caspase-3, RANK/RANKL, ER/PR, CD44, and STAT3/pSTAT3 expression in fallopian tube, ovary, and endometrium; gene expression profiling in the fallopian tube and ovary; serum markers (progesterone, estradiol, C-terminal telopeptide) and denosumab levels; and toxicity. Statistical methods: The primary endpoint is post-treatment Ki67 expression in the fimbrial end of the fallopian tube in the denosumab arm compared to the no treatment arm. Assuming a standard deviation of 5.0%, we will have 82% power to detect a 4.0% absolute difference (or effect size of 0.8) in Ki67 proliferation index between the denosumab and no treatment groups by applying a 2-sample t-","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"20 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"84329638","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-15DOI: 10.1158/1538-7445.SABCS18-OT3-02-06
M. Abu-Khalaf, M. Pierobon, N. Denduluri, F. Valdes-Albini, A. Forero-Torres, A. Clark, R. Yung, M. Mita, S. Christensen, K. Awerkamp, Bryant Dunetz, R. Murphy, C. Hatzis, D. Zelterman, L. Liotta, E. Petricoin
Background: Palbociclib, ribociclib and abemaciclib are 3 cyclin-dependent kinase (CDK) 4/6 inhibitors (inh) approved by the FDA for treatment of patients (pts) with hormone receptor–positive (HR+) metastatic breast cancer (MBC). We hypothesize that measuring the signaling architecture of CDK 4/6 kinase signaling network will predict response to CDK 4/6 inh and identify pts who are unlikely to respond to CDK4/6 inh and can be treated with other FDA approved drugs or a clinical trial. Patients who develop disease progression on CDK 4/6 inh within 12 months of starting therapy are eligible for a unique mulit-omic based molecular analysis that can be used as a therapeutic decision support tool. Trial design: This is an open label, multicenter study prospectively evaluating the phosphoprotein-based CDK 4/6 kinase network within the tumors of 100 pts with HR + MBC who are candidates for standard 1st line treatment with a CDK 4/6 inh plus endocrine therapy (ET). Eligible pts must have pretreatment tissue from a metastatic lesion sufficient to complete baseline biomarker analysis using a novel Laser Capture Microdissecton (LCM) reverse phase protein array (RPPA) coupled approach to quantitatively analyze 8 specific proteins/phosphoproteins within the CDK 4/6 kinase signaling network (Total Rb; phospho Rb (S780); total Cyclin D1; phospho Cyclin D1 (S286); total p16INK; total p27KIP; phosphop27KIP (T187); phosphoFoxM1 (T600). Pts who develop disease progression within 12 months of starting CDK4/6 inh plus ET will be eligible for an optional biopsy of a soft tissue or bone metastatic lesion at time of disease progression for molecular analysis using a multi-omic CAP/CLIA laboratory assays to analyze post-therapy biopsies by RPPA, IHC analysis, RNA-Seq, and targeted exome sequencing. A molecular tumor board then provides the results to the treating physician as a therapeutic decision support tool outlining potential targets and possible therapies which may be used for further treatment. The primary objective is to demonstrate a correlation between one-year progression-free survival (PFS) and pre-treatment phospho RB levels. Secondary endpoints will examine the association between one-year PFS and 7 pre-specified CDK 4/6 signaling network markers. Thirteen of planned 100 patients have been enrolled. The study will be conducted at 12 centers. Clinical trial registry number 03195192 Citation Format: Abu-Khalaf MM, Pierobon M, Denduluri N, Valdes-Albini F, Forero-Torres A, Clark A, Yung R, Mita M, Christensen S, Awerkamp K, Dunetz B, Murphy R, Hatzis C, Zelterman D, Liotta L, Petricoin E. Utilizing multiomic advanced diagnostics to identify CDK4/6 inhibitor response predictors and a post-treatment multiomic signature for patients with ER+/HER2- metastatic breast cancer (SIDEOUT-3) [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT3-02-06.
背景:Palbociclib, ribociclib和abemaciclib是FDA批准用于治疗激素受体阳性(HR+)转移性乳腺癌(MBC)患者的3种细胞周期蛋白依赖性激酶(CDK) 4/6抑制剂(inh)。我们假设测量CDK4/6激酶信号网络的信号结构将预测对CDK4/6的反应,并确定不太可能对CDK4/6反应的患者,可以用其他FDA批准的药物或临床试验进行治疗。在开始治疗的12个月内出现CDK 4/6 / h疾病进展的患者有资格获得独特的基于多组学的分子分析,可作为治疗决策支持工具。试验设计:这是一项开放标签、多中心的研究,前瞻性地评估了100例HR + MBC患者肿瘤中基于磷酸化蛋白的cdk4 /6激酶网络,这些患者是cdk4 /6 inh +内分泌治疗(ET)标准一线治疗的候选人。符合条件的患者必须有足够的转移性病变预处理组织,以完成基线生物标志物分析,使用新型激光捕获显微解剖(LCM)反相蛋白阵列(RPPA)偶联方法定量分析CDK 4/6激酶信号网络中的8种特定蛋白/磷酸化蛋白(Total Rb;磷酸Rb (S780);总Cyclin D1;phospho Cyclin D1 (S286);总p16INK;总p27KIP;phosphop27KIP (T187);phosphoFoxM1 (T600)。在开始CDK4/6 inh + ET治疗后12个月内出现疾病进展的患者,将有资格在疾病进展时选择软组织或骨转移灶活检,进行分子分析,使用多组学CAP/CLIA实验室分析,通过RPPA、IHC分析、RNA-Seq和靶向外显子组测序分析治疗后活检。然后,分子肿瘤委员会将结果提供给治疗医生,作为治疗决策支持工具,概述可能用于进一步治疗的潜在靶点和可能的治疗方法。主要目的是证明一年无进展生存期(PFS)与治疗前磷酸RB水平之间的相关性。次要终点将检查1年PFS与7个预先指定的cdk4 /6信号网络标记物之间的关系。在计划的100名患者中,有13名已经登记。这项研究将在12个中心进行。临床试验注册号03195192引用格式:Abu-Khalaf MM, Pierobon M, Denduluri N, Valdes-Albini F, Forero-Torres A, Clark A, Yung R, Mita M, Christensen S, Awerkamp K, Dunetz B, Murphy R, Hatzis C, Zelterman D, Liotta L, Petricoin E. ER+/HER2-转移性乳腺癌患者CDK4/6抑制剂疗效预测及治疗后多组学特征(SIDEOUT-3)[摘要]。2018年圣安东尼奥乳腺癌研讨会论文集;2018年12月4-8日;费城(PA): AACR;中国癌症杂志,2019;79(4增刊):OT3-02-06。
{"title":"Abstract OT3-02-06: Utilizing multiomic advanced diagnostics to identify CDK4/6 inhibitor response predictors and a post-treatment multiomic signature for patients with ER+/HER2- metastatic breast cancer (SIDEOUT-3)","authors":"M. Abu-Khalaf, M. Pierobon, N. Denduluri, F. Valdes-Albini, A. Forero-Torres, A. Clark, R. Yung, M. Mita, S. Christensen, K. Awerkamp, Bryant Dunetz, R. Murphy, C. Hatzis, D. Zelterman, L. Liotta, E. Petricoin","doi":"10.1158/1538-7445.SABCS18-OT3-02-06","DOIUrl":"https://doi.org/10.1158/1538-7445.SABCS18-OT3-02-06","url":null,"abstract":"Background: Palbociclib, ribociclib and abemaciclib are 3 cyclin-dependent kinase (CDK) 4/6 inhibitors (inh) approved by the FDA for treatment of patients (pts) with hormone receptor–positive (HR+) metastatic breast cancer (MBC). We hypothesize that measuring the signaling architecture of CDK 4/6 kinase signaling network will predict response to CDK 4/6 inh and identify pts who are unlikely to respond to CDK4/6 inh and can be treated with other FDA approved drugs or a clinical trial. Patients who develop disease progression on CDK 4/6 inh within 12 months of starting therapy are eligible for a unique mulit-omic based molecular analysis that can be used as a therapeutic decision support tool. Trial design: This is an open label, multicenter study prospectively evaluating the phosphoprotein-based CDK 4/6 kinase network within the tumors of 100 pts with HR + MBC who are candidates for standard 1st line treatment with a CDK 4/6 inh plus endocrine therapy (ET). Eligible pts must have pretreatment tissue from a metastatic lesion sufficient to complete baseline biomarker analysis using a novel Laser Capture Microdissecton (LCM) reverse phase protein array (RPPA) coupled approach to quantitatively analyze 8 specific proteins/phosphoproteins within the CDK 4/6 kinase signaling network (Total Rb; phospho Rb (S780); total Cyclin D1; phospho Cyclin D1 (S286); total p16INK; total p27KIP; phosphop27KIP (T187); phosphoFoxM1 (T600). Pts who develop disease progression within 12 months of starting CDK4/6 inh plus ET will be eligible for an optional biopsy of a soft tissue or bone metastatic lesion at time of disease progression for molecular analysis using a multi-omic CAP/CLIA laboratory assays to analyze post-therapy biopsies by RPPA, IHC analysis, RNA-Seq, and targeted exome sequencing. A molecular tumor board then provides the results to the treating physician as a therapeutic decision support tool outlining potential targets and possible therapies which may be used for further treatment. The primary objective is to demonstrate a correlation between one-year progression-free survival (PFS) and pre-treatment phospho RB levels. Secondary endpoints will examine the association between one-year PFS and 7 pre-specified CDK 4/6 signaling network markers. Thirteen of planned 100 patients have been enrolled. The study will be conducted at 12 centers. Clinical trial registry number 03195192 Citation Format: Abu-Khalaf MM, Pierobon M, Denduluri N, Valdes-Albini F, Forero-Torres A, Clark A, Yung R, Mita M, Christensen S, Awerkamp K, Dunetz B, Murphy R, Hatzis C, Zelterman D, Liotta L, Petricoin E. Utilizing multiomic advanced diagnostics to identify CDK4/6 inhibitor response predictors and a post-treatment multiomic signature for patients with ER+/HER2- metastatic breast cancer (SIDEOUT-3) [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr OT3-02-06.","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"45 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2019-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89638128","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2018-07-01DOI: 10.1158/1538-7445.AM2018-CT166
Kate Smith, J. Ferrer, Barbara L. Smith, E. Hwang, K. Hunt, Daleela Dodge, S. Karp, S. Valente, I. Wapnir, L. Clark, D. Carr, P. Beitsch, D. Dyess, B. Lesnikoski, P. Blumencranz, N. Dekhne, L. Gold, A. Chagpar, K. Kacena, Livia Gjylameti, F. Geissler
Background: Standard surgical techniques result in positive lumpectomy margins 20-40% of the time. These positive margins require surgical re-excision which places significant burden on the healthcare system and patients. The LUM Imaging System consists of a fluorescence-based imaging agent, a hand-held wide-field detector (LUM Imaging Device) used to image the surgical cavity walls intraoperatively in real-time after the resection of the main lumpectomy specimen, and a proprietary tumor detection algorithm that highlights regions in the tumor bed suspected to contain residual cancer. This imaging system was previously tested in a single-site clinical study. The current study is evaluating the imaging system in a multi-study, large patient cohort. Trial Design / Methods This trial (NCT03321929) is a non-randomized, open-label, multi-site trial designed to further refine the tumor detection algorithm utilized by the LUM Imaging System. This is a prospective, interventional feasibility study and is a pilot arm to a pivotal study which will evaluate the safety and efficacy of the LUM Imaging System. Up to 250 adult female breast cancer patients undergoing lumpectomies are being enrolled at sixteen medical centers across the US. LUM015, a fluorescence-based imaging agent, is injected prior to the subject’s lumpectomy procedure. Surgeons perform their standard of care lumpectomy followed by intraoperative imaging of the lumpectomy cavity with the LUM Imaging System. Specific Aims The primary objective is to assess performance characteristics of the LUM Imaging System and to refine the tumor detection algorithm. A secondary objective is to develop and refine the process of implementing the LUM Imaging System into institution-specific workflows during lumpectomies. Eligibility Criteria This study seeks to enroll women, over the age of 18 and with histologically or cytologically confirmed primary invasive breast cancer (IBC), ductal carcinoma in situ (DCIS) or a combination of IBC/DCIS undergoing a lumpectomy for their breast malignancy. In addition to be willing to follow study procedures, participating in an informed consent discussion, signing an informed consent form, and having baseline lab and screening values within protocol limits, enrolled subjects must meet the following key exclusion criteria: have no history of allergic reaction to polyethylene glycol, no history of allergic reaction to intravenous contrast agents, have not undergone any systemic therapies to treat their cancer, and will not be administered methylene blue or other dye for sentinel lymph node detection during their lumpectomy. Additional detailed eligibility criteria are listed in the protocol. Statistical Methods For categorical variables, summary tabulations of the number and percentage of patients within each category (with a category for missing data) of the parameter will be presented. For continuous variables, the number of patients, mean, median, standard deviation, min
{"title":"Abstract OT3-06-02: Expansion into multiple institutions for training in the use of the LUM Imaging System for intraoperative detection of residual cancer in the tumor bed of female subjects with breast cancer","authors":"Kate Smith, J. Ferrer, Barbara L. Smith, E. Hwang, K. Hunt, Daleela Dodge, S. Karp, S. Valente, I. Wapnir, L. Clark, D. Carr, P. Beitsch, D. Dyess, B. Lesnikoski, P. Blumencranz, N. Dekhne, L. Gold, A. Chagpar, K. Kacena, Livia Gjylameti, F. Geissler","doi":"10.1158/1538-7445.AM2018-CT166","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2018-CT166","url":null,"abstract":"Background: Standard surgical techniques result in positive lumpectomy margins 20-40% of the time. These positive margins require surgical re-excision which places significant burden on the healthcare system and patients. The LUM Imaging System consists of a fluorescence-based imaging agent, a hand-held wide-field detector (LUM Imaging Device) used to image the surgical cavity walls intraoperatively in real-time after the resection of the main lumpectomy specimen, and a proprietary tumor detection algorithm that highlights regions in the tumor bed suspected to contain residual cancer. This imaging system was previously tested in a single-site clinical study. The current study is evaluating the imaging system in a multi-study, large patient cohort. Trial Design / Methods This trial (NCT03321929) is a non-randomized, open-label, multi-site trial designed to further refine the tumor detection algorithm utilized by the LUM Imaging System. This is a prospective, interventional feasibility study and is a pilot arm to a pivotal study which will evaluate the safety and efficacy of the LUM Imaging System. Up to 250 adult female breast cancer patients undergoing lumpectomies are being enrolled at sixteen medical centers across the US. LUM015, a fluorescence-based imaging agent, is injected prior to the subject’s lumpectomy procedure. Surgeons perform their standard of care lumpectomy followed by intraoperative imaging of the lumpectomy cavity with the LUM Imaging System. Specific Aims The primary objective is to assess performance characteristics of the LUM Imaging System and to refine the tumor detection algorithm. A secondary objective is to develop and refine the process of implementing the LUM Imaging System into institution-specific workflows during lumpectomies. Eligibility Criteria This study seeks to enroll women, over the age of 18 and with histologically or cytologically confirmed primary invasive breast cancer (IBC), ductal carcinoma in situ (DCIS) or a combination of IBC/DCIS undergoing a lumpectomy for their breast malignancy. In addition to be willing to follow study procedures, participating in an informed consent discussion, signing an informed consent form, and having baseline lab and screening values within protocol limits, enrolled subjects must meet the following key exclusion criteria: have no history of allergic reaction to polyethylene glycol, no history of allergic reaction to intravenous contrast agents, have not undergone any systemic therapies to treat their cancer, and will not be administered methylene blue or other dye for sentinel lymph node detection during their lumpectomy. Additional detailed eligibility criteria are listed in the protocol. Statistical Methods For categorical variables, summary tabulations of the number and percentage of patients within each category (with a category for missing data) of the parameter will be presented. For continuous variables, the number of patients, mean, median, standard deviation, min","PeriodicalId":19476,"journal":{"name":"Ongoing Clinical Trials","volume":"25 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83372229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}