Dose optimization

剂量优化
  • 文章类型: Case Reports
    银屑病是一种主要影响皮肤的慢性免疫介导的炎症性全身性疾病。Secukinumab是一种被批准用于治疗中度至重度银屑病的抗白细胞介素-17A药物,在临床试验和现实实践中都研究了疗效和安全性。在银屑病成人的维持反应阶段批准的Secukinumab数据表剂量为每月300mg。最近,已经提出了牛皮癣生物疗法的剂量优化,以个性化治疗,寻求更好的治疗依从性和更好的成本效益,因为它们的高成本。在这种情况下,我们报告了我们在中重度银屑病患者接受Secukinumab治疗的剂量优化方面的实际经验.
    Psoriasis is a chronic immune-mediated inflammatory systemic disease that mainly affects skin. Secukinumab is an anti-interleukin-17A agent approved for the treatment of moderate-to-severe psoriasis that has probed efficacy and safety both in clinical trials and in real-world practice. Secukinumab data sheet dose approved in maintenance response phase for adults with psoriasis is 300mg monthly. Recently, dose optimization for biologic therapies in psoriasis has been proposed in order to individualise the treatment looking for better treatment adherence and better cost-effectiveness due to their high costs. In this scenario, we report our real-world experience in dose optimization for patients with moderate-to-severe psoriasis treated with Secukinumab.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    计算机断层扫描(CT)中的剂量优化至关重要,特别是在用于实时导航的CT透视(荧光CT)中,影响患者和操作者的安全。这项研究评估了使用锡过滤器(Sn过滤器)的光谱X射线过滤的影响,和一种称为部分角度计算机断层扫描(PACT)的方法,这涉及在介入放射科医生(IR)位置以环境剂量率H**(10)分段关闭X射线管电流。使用120kVX射线管电压在两个身体区域(上身:头/颈;下身:小腿/脚)进行测量,3×5.0mmCT准直,0.5s转速,和43Eff的X射线管电流。mAs(无Sn滤波器)和165Eff。mAs(带Sn滤波器)。研究发现,当同时使用Sn过滤器和PACT时,两个身体区域的剂量均显着减少。例如,在上身区域,与未使用这些功能的方案相比,组合方案将H•*(10)从11.8µSv/s降低至6.1µSv/s(p<0.0001).约8%的减少(约0.5µSv/s)归因于Sn过滤器(p=0.0005)。这种方法表明,使用Sn滤波器和PACT可以有效地最大程度地减少IR的辐射暴露,特别是保护头部/颈部等区域,只能用(标准)辐射防护材料覆盖不充分。
    Dose optimization in computed tomography (CT) is crucial, especially in CT fluoroscopy (fluoro-CT) used for real-time navigation, affecting both patient and operator safety. This study evaluated the impact of spectral X-ray filtering using a tin filter (Sn filter), and a method called partial-angle computed tomography (PACT), which involves segmentally switching off the X-ray tube current at the ambient dose rate H˙*(10) at the interventional radiologist\'s (IR) position. Measurements were taken at two body regions (upper body: head/neck; lower body: lower legs/feet) using a 120 kV X-ray tube voltage, 3 × 5.0 mm CT collimation, 0.5 s rotation speed, and X-ray tube currents of 43 Eff.mAs (without Sn filter) and 165 Eff.mAs (with Sn filter). The study found significant dose reductions in both body regions when using the Sn filter and PACT together. For instance, in the upper body region, the combination protocol reduced H˙*(10) from 11.8 µSv/s to 6.1 µSv/s (p < 0.0001) compared to the protocol without using these features. Around 8% of the reduction (about 0.5 µSv/s) is attributed to the Sn filter (p = 0.0005). This approach demonstrates that using the Sn filter along with PACT effectively minimizes radiation exposure for the IR, particularly protecting areas like the head/neck, which can only be insufficiently covered by (standard) radiation protection material.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:描述间歇性碳青霉烯输注的护士准备和给药。
    方法:这项观察性研究记录了在三个普通重症监护病房中对成年患者的碳青霉烯输注过程。
    方法:观察输注时间和持续时间。进行输注项目的体积分析以确定在制备和施用阶段重建的碳青霉烯的损失。
    结果:观察到对20名成年患者进行碳青霉烯输注(n=223)。输注持续时间指导是可变的,有两个ICU遵循当前的文献建议,和一个ICU参考药物包装插入信息。在这些参数中,只有60%的输液符合输液持续时间.不遵守计划的给药时间会影响所需的给药间隔。在以下过程中发现了预期剂量的不完全输送:小瓶的次优重建,重建的小瓶数量不正确,未能施用剂量(错过剂量),丢弃输液物品中的抗生素残留物。输注项目的体积分析显示在丢弃的小瓶和注射器中平均剂量损失4.9%和1.2%。在废弃的输液袋和输液管路上分别发生6.3%和30.8%的平均药物损失。没有观察到冲洗指导或实践。
    结论:不正确的护士使用抗生素会导致不同的输注持续时间和未给药处方剂量。给药不足有可能导致细菌抗生素抗性的选择压力。通过输注静脉内递送抗微生物剂的频率增加需要了解所需的施用持续时间以及一旦输注完成如何管理静脉内管线中剩余的残留药物。
    结论:间歇性抗菌输注后冲洗给药线并不常见。尽管重症监护领域存在抗菌药物耐药性的多因素风险因素,护士输液实践必须确保患者接受预期的抗菌治疗.必须注意在处置含有未递送的抗微生物药物的输液物品时,由于环境污染而产生抗微生物剂耐药性的可能性。
    OBJECTIVE: To describe nurse preparation and administration of intermittent carbapenem infusions.
    METHODS: This observational study documented the carbapenem infusion process to adult patients in three general intensive care units.
    METHODS: Timing and duration of infusions were observed. Volumetric analysis of infusion items was conducted to determine loss of reconstituted carbapenem during preparation and administration phases.
    RESULTS: Carbapenem infusions (n = 223) administered to twenty adult patients were observed. Infusion duration guidance was variable, with two ICUs following current literature recommendations, and one ICU referring to medication package insert information. Within these parameters, only 60 % of infusions complied with infusion duration. Non-compliance with planned time of administration impacted on desired dosing intervals. Incomplete delivery of intended dose was found during: sub-optimal reconstitution of vials, incorrect number of vials reconstituted, failure to administer a dose (missed dose), and discarding antibiotic residue in infusion items. Volumetric analysis of infusion items showed mean dose losses of 4.9 % and 1.2 % in discarded vials and syringes. Mean drug losses of 6.3 % and 30.8 % occurred in discarded infusion bags and infusion lines respectively. No flushing guidance or practice was observed.
    CONCLUSIONS: Incorrect nurse administration of antibiotics resulted in varying durations of infusions and the non-delivery of prescribed dose. Under-dosing has the potential to contribute to selection pressure for bacterial antibiotic resistance. The increasing frequency of intravenous delivery of antimicrobial agents through infusions requires an understanding of the required duration of administration and how to manage residual drug remaining in the intravenous line once the infusion is completed.
    CONCLUSIONS: Flushing of administration lines is not common practice following intermittent antimicrobial infusions. Although there are multi-factorial risk factors for antimicrobial resistance in the critical care arena, nurse infusion practice must ensure that patients receive intended antimicrobial treatment. Attention must be given to the potential for antimicrobial resistance from environmental contamination with the disposal of infusion items containing undelivered antimicrobial medication.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    氯雷他定代谢为地氯雷他定。两者都已用于儿童过敏治疗。解剖学,生理,儿童的生物学参数和药物清除率随年龄而变化。我们旨在开发基于生理的全身药代动力学(PBPK)模型,以同时预测儿童中氯雷他定和地氯雷他定的药代动力学。在使用11个成人数据集进行验证后,将开发的PBPK模型外推到儿童.对不同年龄儿童口服氯雷他定或地氯雷他定后的血浆浓度进行了模拟,并与六个儿童数据集进行了比较。缩放解剖学/生理学后,蛋白质结合,和间隙,令人满意地预测了这两种药物在儿科人群中的药代动力学。在1000个虚拟儿童中,大多数观察到的浓度都落在模拟的第5-95百分位数范围内。浓度-时间曲线下的预测面积(AUC)和Cmax落在观测值的0.5-2.0倍范围内。在成人服用10mg氯雷他定或5mg地氯雷他定后,根据相似的AUC模拟了不同年龄儿童的氯雷他定或地氯雷他定的口服剂量。成功开发了小儿PBPK模型,可同时预测所有年龄段儿童的氯雷他定和地氯雷他定的血浆浓度。开发的儿科PBPK模型也可用于优化儿科剂量。
    Loratadine is metabolized to desloratadine. Both of them have been used for allergy treatment in children. Anatomical, physiological, and biological parameters of children and clearance of drugs vary with age. We aimed to develop a whole-body physiologically based pharmacokinetic (PBPK) model to simultaneously predict the pharmacokinetics of loratadine and desloratadine in children. Following validation using 11 adult data sets, the developed PBPK model was extrapolated to children. Plasma concentrations following oral loratadine or desloratadine to children of different ages were simulated and compared with six children data sets. After scaling anatomy/physiology, protein binding, and clearance, pharmacokinetics of the two drugs in pediatric populations were satisfactorily predicted. Most of the observed concentrations fell within the 5th-95th percentile range of the simulations in 1000 virtual children. The predicted area under the concentration-time curve (AUC) and Cmax fell within 0.5-2.0-fold range of the observations. Oral doses of loratadine or desloratadine for children of different ages were simulated based on similar AUCs following 10 mg of loratadine or 5 mg of desloratadine for adults. Pediatric PBPK model was successfully developed to simultaneously predict plasma concentrations of loratadine and desloratadine in children of all ages. The developed pediatric PBPK model may also be applied to optimize pediatric dosage.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    剂量优化是药物开发中的关键挑战。历史上,由于肿瘤学的独特特征和要求,肿瘤学的剂量确定与其他非肿瘤学治疗领域有着不同的路径。然而,随着新的药物模式和肿瘤学药物机制的出现,比如免疫疗法,放射性药物,靶向治疗,细胞抑制剂,和其他人,与细胞毒性化疗相比,疗效和毒性的剂量-反应关系可能有很大差异.低于MTD的剂量可能表现出与MTD相似的功效,具有改善的耐受性。类似于在非肿瘤治疗中通常观察到的。因此,肿瘤药物开发的新模式需要剂量优化的替代策略.本文探讨了从非肿瘤学到肿瘤学的剂量发现方法的历史演变,突出例子并总结变化的潜在驱动因素。随后,提供了实用的框架和指导,以说明如何将剂量优化纳入开发计划的各个阶段。我们提供以下一般建议:1)第一阶段的目标是确定剂量范围而不是单个MTD剂量,以便随后开发,以更好地表征剂量范围内的安全性和耐受性。2)推荐至少两种可通过PK分离的剂量用于II期的剂量优化。3)理想情况下,在开展验证性研究之前,应进行剂量优化。然而,创新设计,如无缝II/III设计可以用于剂量选择,并可能加速药物开发计划。
    Dose optimization is a critical challenge in drug development. Historically, dose determination in oncology has followed a divergent path from other non-oncology therapeutic areas due to the unique characteristics and requirements in Oncology. However, with the emergence of new drug modalities and mechanisms of drugs in oncology, such as immune therapies, radiopharmaceuticals, targeted therapies, cytostatic agents, and others, the dose-response relationship for efficacy and toxicity could be vastly varied compared to the cytotoxic chemotherapies. The doses below the MTD may demonstrate similar efficacy to the MTD with an improved tolerability profile, resembling what is commonly observed in non-oncology treatments. Hence, alternate strategies for dose optimization are required for new modalities in oncology drug development. This paper delves into the historical evolution of dose finding methods from non-oncology to oncology, highlighting examples and summarizing the underlying drivers of change. Subsequently, a practical framework and guidance are provided to illustrate how dose optimization can be incorporated into various stages of the development program. We provide the following general recommendations: 1) The objective for phase I is to identify a dose range rather than a single MTD dose for subsequent development to better characterize the safety and tolerability profile within the dose range. 2) At least two doses separable by PK are recommended for dose optimization in phase II. 3) Ideally, dose optimization should be performed before launching the confirmatory study. Nevertheless, innovative designs such as seamless II/III design can be implemented for dose selection and may accelerate the drug development program.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:提供儿科介入心脏病学手术中的辐射暴露数据,解决有价值的本地诊断参考水平(LDRL)的稀缺问题,根据辐射防护185报告(RP185)提出的标准化方法建立。
    方法:2019年9月至2022年12月在帕多瓦大学医院进行的儿科导尿程序按体重(BW)类别和程序类型进行分层。计算LDRL为至少20名患者的第75百分位数的Kerma面积乘积(PKA)和参考点的AirKerma(Ka,r)值。应用Kruskal-Wallis检验来评估所选程序的BW组之间和相同BW等级的程序之间的剂量相关量的差异。将结果与最近的文献进行比较。
    结果:共分析了838例手术。为五个治疗程序提供了LDRL。PKA和Ka的第75百分位数,r随体重增加,无论程序类型。PKA和Ka,R通常在BW组之间有统计学差异,对于诊断和治疗程序,在固定体重组的不同程序之间。血管成形术和右心室流出道治疗(PVR)显示暴露值大约是其他程序的两倍。除房间隔缺损(ASD)闭合外,PKA/(BW·FT)在程序之间没有统计学差异。这项研究的LDRL值通常低于已发表的值。
    结论:该研究是少数针对体重类别和手术类型提供大量LDRL的研究之一,每组至少有20名患者。与RP185达成协议。PKA与产品BW·FT具有很强的相关性。
    OBJECTIVE: To provide data on radiation exposure in paediatric interventional cardiology procedures, addressing the scarcity of valuable Local Diagnostic Reference Levels (LDRLs),established according to the standardized approach proposed by the Radiation Protection 185 report (RP185).
    METHODS: Paediatric catheterization procedures conducted at the University-Hospital of Padua from September 2019 to December 2022 were stratified by body weight (BW) classes and procedure type. LDRLs were calculated for groups with at least 20 patients as the 75th percentile of Kerma-Area Product (PKA) and Air Kerma at reference point (Ka,r) values. Kruskal-Wallis test was applied to evaluate differences in the dose-related quantities among BW groups for a selected procedure and among procedures for the same BW class. Results were compared with recent literature.
    RESULTS: A total of 838 procedures were analysed. LDRL were provided for five therapeutic procedures. The 75th percentile of PKA and Ka,r increases with weight, regardless procedure type. PKA and Ka,r are generally statistically different between BW groups, for both diagnostic and therapeutic procedures, and between different procedures at fixed weight group. Angioplasty and Right Ventricular Outflow Tract treatments (PVR) showed exposure values approximately doubled then other procedures. PKA/(BW·FT) is not statistically different among procedures except for Atrial Septal Defect (ASD) closures. LDRL values from this study are generally lower than the published ones.
    CONCLUSIONS: The study stands out as one of the few that presents a considerable number of LDRLs for weight categories and procedure types with a sample size of at least 20 patients per group, in agreement with RP185. PKA shows strong correlation with the product BW·FT.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    用于肿瘤学登记试验的传统剂量选择通常采用一步或两步单一最大耐受剂量(MTD)方法。然而,这种方法可能不适合分子靶向治疗,其往往具有与细胞毒性剂明显不同的毒性特征。美国食品和药物管理局启动了Optimus项目,以改革肿瘤药物开发中的剂量优化,并在最近发布了相关的行业指南。
    我们提出了“剂量优化的三个步骤”程序,为了响应这些倡议,并讨论剂量优化设计和分析中的细节。第一步是剂量递增,以通过有效的混合设计确定MTD或最大给药剂量。这可以提供良好的过量控制,并增加推荐的MTD接近真实MTD的可能性。第二步是选择适当的推荐剂量进行扩展(RDE),基于所有可用的数据,包括新兴的安全,药代动力学,药效学,和其他生物标志物信息。第三步是剂量优化,它使用来自随机分数阶乘设计的数据,在扩展阶段在多个肿瘤队列中探索多个RDE,以确保为注册试验选择可行的剂量,并确定对研究治疗最敏感的肿瘤类型。
    我们相信使用这种三步方法可以增加为注册试验选择最佳剂量的可能性,该试验证明了平衡的安全性,同时保留了在MTD中观察到的大部分功效。
    UNASSIGNED: Traditional dose selection for oncology registration trials typically employs a one- or two-step single maximum tolerated dose (MTD) approach. However, this approach may not be appropriate for molecularly targeted therapy, which tends to have toxicity profiles that are markedly different than cytotoxic agents. The US Food and Drug Administration launched Project Optimus to reform dose optimization in oncology drug development and has recently released a related guidance for industry.
    UNASSIGNED: We propose a \"three steps toward dose optimization\" procedure, in response to these initiatives, and discuss the details in dose-optimization designs and analyses. The first step is dose escalation to identify the MTD or maximum administered dose with an efficient hybrid design, which can offer good overdose control and increases the likelihood of the recommended MTD being close to the true MTD. The second step is the selection of appropriate recommended doses for expansion (RDEs), based on all available data, including emerging safety, pharmacokinetics, pharmacodynamics, and other biomarker information. The third step is dose optimization, which uses data from a randomized fractional factorial design with multiple RDEs explored in multiple tumor cohorts during the expansion phase to ensure a feasible dose is selected for registration trials, and that the tumor type most sensitive to the investigative treatment is identified.
    UNASSIGNED: We believe using this three-step approach can increase the likelihood of selecting an optimal dose for a registration trial that demonstrates a balanced safety profile while retaining much of the efficacy observed at the MTD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    败血症的有效治疗不仅需要及时施用适当的抗微生物剂,而且需要精确的剂量以提高患者存活的可能性。适当的给药是指在感染部位产生治疗药物浓度的剂量的施用。这确保了有利的临床和微生物学反应,同时避免了抗生素相关的毒性。治疗药物监测(TDM)是实现这些目标的推荐方法。然而,TDM并非在所有重症监护病房(ICU)和所有抗微生物剂中普遍可用。在没有TDM的情况下,医疗保健从业者需要依靠几个因素来做出明智的剂量决定。这些包括患者的临床状况,致病病原体,器官功能障碍的影响(需要体外治疗),和抗菌剂的物理化学性质。在这种情况下,在ICU住院期间,不同危重患者和同一患者体内的抗微生物药物的药代动力学差异很大.这种可变性强调了对个体化给药的需要。这篇综述旨在描述在危重患者中观察到的主要病理生理变化及其对抗菌药物剂量决定的影响。它还旨在提供必要的实用建议,以帮助临床医生优化重症患者的抗菌治疗。
    Effective treatment of sepsis not only demands prompt administration of appropriate antimicrobials but also requires precise dosing to enhance the likelihood of patient survival. Adequate dosing refers to the administration of doses that yield therapeutic drug concentrations at the infection site. This ensures a favorable clinical and microbiological response while avoiding antibiotic-related toxicity. Therapeutic drug monitoring (TDM) is the recommended approach for attaining these goals. However, TDM is not universally available in all intensive care units (ICUs) and for all antimicrobial agents. In the absence of TDM, healthcare practitioners need to rely on several factors to make informed dosing decisions. These include the patient\'s clinical condition, causative pathogen, impact of organ dysfunction (requiring extracorporeal therapies), and physicochemical properties of the antimicrobials. In this context, the pharmacokinetics of antimicrobials vary considerably between different critically ill patients and within the same patient over the course of ICU stay. This variability underscores the need for individualized dosing. This review aimed to describe the main pathophysiological changes observed in critically ill patients and their impact on antimicrobial drug dosing decisions. It also aimed to provide essential practical recommendations that may aid clinicians in optimizing antimicrobial therapy among critically ill patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    对于75岁以上的老年患者,在全身麻醉期间是否需要调整静脉注射利多卡因的剂量尚不清楚。本研究旨在探讨年龄对全身麻醉患者静脉注射利多卡因药代动力学(PK)和安全性的影响。从76名全身麻醉患者中收集了599份血浆样本,这些患者分为三个年龄组:18-64岁、65-74岁和≥75岁。利多卡因以1.5mg/kg的剂量静脉内给药18-64岁和65-74岁组,而≥75岁组的剂量调整为1.0mg/kg。使用经过验证的超高效液相色谱-串联质谱法测定利多卡因及其活性代谢物的血浆浓度,数据采用非房室分析进行分析.结果表明,利多卡因及其代谢物的PK没有明显的年龄相关差异。在三个年龄组中,当剂量标准化为1.5mg/kg时,超过90%的患者利多卡因浓度在安全有效范围内.总之,对于86岁以下接受全身麻醉的患者,静脉注射利多卡因不需要根据年龄调整剂量.
    It remains unclear whether dosage adjustment of intravenous lidocaine is necessary during general anesthesia for elderly patients over 75 years old. This study aimed to investigate the effects of age on the pharmacokinetics (PK) and safety of intravenous lidocaine in patients undergoing general anesthesia. A total of 599 plasma samples were collected from 76 general anesthesia patients across three age groups: 18-64, 65-74, and ≥ 75 years. Lidocaine was administered intravenously at a dose of 1.5 mg/kg for the 18-64 and 65-74 years groups, while the dose was adjusted to 1.0 mg/kg for the ≥ 75 years group. The plasma concentrations of lidocaine and its active metabolites were measured using a validated ultra-performance liquid chromatography-tandem mass spectrometry assay, and the data were analyzed using a noncompartmental analysis. The results revealed no significant age-related differences in the PK of lidocaine and its metabolites. Among the three age groups, over 90 % of patient achieved a lidocaine concentration within a safe and effective range when the dosage was normalized to 1.5 mg/kg. In conclusion, age-based dosage adjustment was unnecessary for intravenous lidocaine in patients below 86 years undergoing general anesthesia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肿瘤药物开发早期的剂量选择和优化是后期药物开发成功的基础。双变量贝叶斯逻辑回归模型(BLRM)是一种广泛使用的基于模型的算法,已被证明可以提高基于剂量限制毒性(DLT)识别推荐的2期剂量(RP2D)的准确性传统方法,例如3+3。然而,在I期试验的升级和扩展阶段,优化剂量选择以在安全性和有效性之间取得适当平衡仍然是一个挑战.在本文中,我们首先使用一项I期临床试验,以证明试验参与者中与药代动力学(PK)参数相关的药物暴露的变异性可能会增加确定最佳剂量的困难.我们使用模拟来表明,同时或回顾性地拟合BLRM模型,以剂量无关的PK参数作为协变量,可以提高识别DLT速率在预定毒性区间内的剂量水平的准确性。此外,我们提出了基于模型和规则的方法,根据PK/暴露参数在扩展队列中修改患者水平的剂量.模拟研究表明,这种方法导致在I期试验的扩展阶段进行筛选后,具有可控毒性和理想疗效裕度的剂量水平被推进到后期管道的可能性更高。
    Dose selection and optimization in early phase of oncology drug development serves as the foundation for the success of late phases drug development. Bivariate Bayesian logistic regression model (BLRM) is a widely utilized model-based algorithm that has been shown to improve the accuracy for identifying recommended phase 2 dose (RP2D) based on dose-limiting-toxicity (DLT) over traditional method such as 3 + 3. However, it remains a challenge to optimize dose selection that strikes a proper balance between safety and efficacy in escalation and expansion phase of phase I trials. In this paper, we first use a phase I clinical trial to demonstrate how the variability of drug exposure related to pharmacokinetic (PK) parameters among trial participants may add to the difficulties of identifying optimal dose. We use simulation to show that concurrently or retrospectively fitting BLRM model for dose/toxicity data from escalation phase with dose-independent PK parameters as covariate lead to improved accuracy of identifying dose level at which DLT rate is within a prespecified toxicity interval. Furthermore, we proposed both model- and rule-based methods to modify dose at patient level in expansion cohorts based on their PK/exposure parameters. Simulation studies show this approach leads to higher likelihood for a dose level with a manageable toxicity and desirable efficacy margin to be advanced to late phase pipeline after being screened at expansion phase of phase I trial.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号