跳转到主要内容
主页 / 资源 / 博客 / 机制性建模加速双特异性(和多特异性)抗体 (bsAbs) 药物开发的4 种方法

机制性建模加速双特异性(和多特异性)抗体 (bsAbs) 药物开发的4 种方法

With nine FDA-approved drugs today, 100+ in clinical development and several hundred in the preclinical stage, the market for bsAbs is projected to grow to >$30B by 2028. The distinct advantages of bsAbs, such as improved selectivity and specificity, increased efficacy, and lower toxicity, will result in measurable benefits for cancer patients. But bsAbs are not the same as combination therapy and require a greater understanding of the drug (s) and biological system being targeted.

This work requires the use of a mechanistic modeling approach, specifically Quantitative Systems Pharmacology (QSP), which benefit drug developers in 4 ways:

  1. Selection of starting dose: getting to IND approval faster and at lower cost.
  2. Defining efficacious dose: sources of variability and mechanism of action of two antibodies is not additive, so traditional dose escalation methods often fail.
  3. Managing safety concerns: reduce the impact of cytokine release syndrome (CRS) via modeling.
  4. Predicting and managing between patient variability: using virtual twins to amplify patient characteristics and differentiation to employ the best therapeutic approach.

BsAbs: Advantages and Opportunities for Revolutionizing the field of Immuno-oncology

As opposed to a monoclonal antibody, in which both halves” of the Y-shaped molecule are identical and bind to the same antigen target, the two halves of the bsAbs antibody are different, allowing simultaneous binding to two different targets. These may be antigens on two different cells, or two different antigens on the same cell. By targeting two antigens or epitopes, bsAbs can deliver multiple physiological and anti-tumor responses. As opposed to combination therapy, the two mAbs may work independently or in connection with one another, but patients only need to take a single treatment. The synergistic features of the bsAbs enable a wider range of therapeutic applications and are propelled by the growth in cancer patients and types, increased demand for targeted therapies, improved efficacy and safety profiles, and desire for personalized medicine.

Already in development, the next generation of targeted therapies are multispecifics, protein-based therapeutic molecules that can bind to more than two targets simultaneously.



The Challenges in BsAbs Development

While of great therapeutic potential, bsAbs (and multispecifics) are complex modalities with drug development challenges to overcome including:

  • Selection of optimal targets and modalities appropriate for the mechanism of action.
  • Design of bsAbs including optimal affinities for each target.
  • Mechanistic complexities such as role of avidity and the bell-shaped concentration response relationship.
  • Selection of relevant starting doses using a minimal anticipated biological effect level.
  • Predicting efficacious dose despite nonintuitive dose response relationships.
  • Navigating efficacy vs. toxicity.

A unique issue for bsAbs relates to dose prediction, as per the seminal tutorial from Betts and van der Graaf1. “Historically, in oncology drug development, efficacy has been assumed to be dose related and cancer drugs are escalated to the maximum tolerated dose.”The complex mechanism of action of BsAbs can result in nonlinear relationships between dose and trimer exposure, resulting in a bell- shaped concentration vs response curve as shown in the diagram.



Mechanistic, QSP modeling approaches are a powerful integrative tool to understand the complexities and aid in bsAb design and optimization, clinical translation, trial design, and prediction of regimens and strategies to reduce dose limiting toxicities of bsAbs. QSP modeling is increasingly used to support regulatory (IND) submissions.

Case studies Affirm Value of QSP in Translational Stages of bsAbs and multi-specific development

  1. Selection of starting dose: The sponsor had two bsAb molecules moving toward IND submittal in a six-month period. The first followed the traditional MABEL approach, the second replaced FIH prediction using a QSP model. The QSP model enabled the sponsor to save two full dose levels, attaining the IND faster and at lower cost.
  2. Defining efficacious dose: In this example for a T cell engager bsAb, the QSP model was able to accurately predict the observed clinical efficacy dose range when the dose was estimated based on in vivo-data derived nACT (TCE and tumor cell normalized to number of tumor cells (nACT).
  3. Managing safety concerns: A common side effect of bsAb immunotherapy is cytokine release syndrome (CRS), a potentially life-threatening complication in which activated immune cells release a large number of cytokines into the bloodstream, resulting in systemic inflammation. In this case example, the QSP model was used to assess the translatability of preclinical data and predicted that the molecule could achieve efficacy without compromising cytotoxic activity on human tumors to cross-reactivity considerations.
  4. Predicting and managing between patient variability. Certara’s Virtual Twin technology creates a computer-simulated model of each patient, replicating the patient’s various attributes that affect a drug’s fate in their body and hence its effects. This mechanistic modeling approach can predict the optimal drug dosing regimen for an individual patient – one that maximizes therapeutic benefit while minimizing side effects – by evaluating the impact of different drug doses, schedules, and combinations in the patients’ in silico ‘virtual twin’ first.

Certara Simcyp has developed a platform for mechanistic modeling of bsAbs using our QSP Designer software, which was introduced via this poster paper at the September 2023 Annual Consortium meeting. With >20 completed bsAb and multi-specifics projects and a PBPK/QSP platform, we can perform a FIH dose prediction project in less than two months.

Betts, A and van der Graaf, P., “Mechanistic Quantitative Pharmacology Strategies for the Early Clinical Development of Bispecific Antibodies in Oncology,” Clin Pharmacol Ther. 2020 Sep;108(3):528-541.

关于作者

Piet van der Graaf, PharmD, PhD
By: Piet van der Graaf, PharmD, PhD

Piet van der Graaf 现任 Certara 高级副总裁兼 QSP 负责人,同时也是莱顿大学系统药理学教授。Piet 是 2013 - 2016 年莱顿药物研究学术中心的研究主任。1999 - 2013 年,Piet 在辉瑞公司的研发生物学、药代动力学和药物代谢、以及临床药理学等部门担任过多种领导职务。Piet 于2012 - 2018 年担任 CPT: Pharmacometrics & Systems Pharmacology 的创始主编,随后成为 Clinical Pharmacology & Therapeutics 的主编。Piet 曾在伦敦国王学院师从诺贝尔奖获得者 Sir James Black,接受了临床医学的博士培训。他曾获得 2024 年美国临床药理学与治疗学会(ASCPT)颁发的 Gary Neil 药物开发创新奖,同时也是国际定量药理学会(ISoP)2021 年领袖奖的获得者。Piet 是英国药理学会的当选委员,在定量药理学和药物开发领域发表了超过 200 篇同行评议的论文。

沪ICP备2022021526号

Powered by Translations.com GlobalLink OneLink Software