scholarly journals Cardiovascular Safety Assessment in Cancer Drug Development

Author(s):  
Ohad Oren ◽  
Tomas G. Neilan ◽  
Michael G. Fradley ◽  
Deepak L. Bhatt

Abstract The development of cardiovascular toxicity attributable to anticancer drugs is a pivotal event that is associated with cardiovascular morbidity as well as with worse cancer‐specific and overall outcomes. Although broad consensus exists regarding the importance of cardiovascular safety assessment in cancer drug development, real‐world data suggest that cardiovascular events are significantly underestimated in oncology trials. This drug safety discrepancy has profound implications on drug development decisions, risk‐benefit evaluation, formulation of surveillance and prevention protocols, and survivorship. In this article, we review the contemporary cardiovascular safety evaluation of new pharmaceuticals in hematology and oncology, spanning from in vitro pharmacodynamic testing to randomized clinical trials. We argue that cardiovascular safety assessment of anticancer drugs should be reformed and propose practical strategies, including development and validation of preclinical assays, expansion of oncology trial eligibility, incorporation of cardiovascular end points in early‐phase studies, and design of longitudinal multi‐institutional cardiotoxicity registries.

Author(s):  
Lauren Marshall ◽  
Isabel Löwstedt ◽  
Paul Gatenholm ◽  
Joel Berry

The objective of this study was to create 3D engineered tissue models to accelerate identification of safe and efficacious breast cancer drug therapies. It is expected that this platform will dramatically reduce the time and costs associated with development and regulatory approval of anti-cancer therapies, currently a multi-billion dollar endeavor [1]. Existing two-dimensional (2D) in vitro and in vivo animal studies required for identification of effective cancer therapies account for much of the high costs of anti-cancer medications and health insurance premiums borne by patients, many of whom cannot afford it. An emerging paradigm in pharmaceutical drug development is the use of three-dimensional (3D) cell/biomaterial models that will accurately screen novel therapeutic compounds, repurpose existing compounds and terminate ineffective ones. In particular, identification of effective chemotherapies for breast cancer are anticipated to occur more quickly in 3D in vitro models than 2D in vitro environments and in vivo animal models, neither of which accurately mimic natural human tumor environments [2]. Moreover, these 3D models can be multi-cellular and designed with extracellular matrix (ECM) function and mechanical properties similar to that of natural in vivo cancer environments [3].


2018 ◽  
Vol 08 (03) ◽  
Author(s):  
Laura Vidal Boixader ◽  
Kelly Kevelin Cartis ◽  
Liat Vidal ◽  
Jozsef Palatka ◽  
Jim Wahl ◽  
...  

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e13572-e13572
Author(s):  
Sundeep Agrawal ◽  
Shaily Arora ◽  
Jonathon Joseph Vallejo ◽  
Thomas Gwise ◽  
Meredith Kathleen Chuk ◽  
...  

e13572 Background: Improved understanding of the underlying biology of cancer has led to a paradigm shift in cancer drug development and has paved the way for many products to receive accelerated or regular approval based on non-randomized/single arm trials (SATs). Given the high unmet medical need of cancer patients, challenges with lengthy and confounded survival endpoints, and difficulty enrolling rare biomarker-defined subsets of disease, SATs have been used to evaluate a variety of cancer therapies. Unlike time to event endpoints, the objective and clinically relevant endpoint of response rate (RR) and duration of response is interpretable in SATs, as spontaneous tumor shrinkage is not expected. Methods: A search of FDA databases identified all drugs and biologics approved for malignant hematology and oncology indications from January 1, 2001, to December 31, 2020 based on SATs. Data sources included approval letters, U.S. prescribing information, and clinical review documents. The definition of response varied by setting and time period (e.g. RECIST, WHO, IWG, etc.). Results: Between January 1, 2001 and December 31, 2020, FDA granted 153 new indications based on SATs, including 102 accelerated approvals (AAs) and 51 regular approvals (RAs). Overall, 69 approvals (45%) were for new molecular entities and 84 (55%) were expanded indications. Response rate was the most common endpoint used in the trial providing substantial evidence of efficacy to support approval [120/153, (78%)]. The durability of response was also considered to support evidence of clinical benefit. Of the 102 AAs, 38 (37%) have fulfilled their post-marketing requirement (PMR) to verify clinical benefit, 59 (58%) are pending verification of benefit, and 5 (5%) have been withdrawn from the market. Of note, 88% (52/59) of AAs pending verification of benefit occurred in the last 5 years alone (22 AAs in 2020, 8 in 2019, 8 in 2018, 12 in 2017, and 2 in 2016). Between 2001-2020, 58 (38%) new indications were granted for kinase inhibitors, 34 (22%) for immune checkpoint inhibitors (CPIs), and 61 (40%) for drugs with other mechanisms of action including but not limited to antibody-drug conjugates, cytotoxic drugs, and non-CPI monoclonal antibodies. Conclusions: In the last two decades, SATs have been effectively used to study anti-cancer therapies in well-defined patient populations using durable RR as an objective and interpretable clinical endpoint. Although randomized clinical trials remain the gold standard in clinical research, SATs have allowed for rapid advancements in oncology drug development and will continue to serve an important role in bringing new therapies to patients with unmet need.


2018 ◽  
Vol 101 ◽  
pp. 69-76 ◽  
Author(s):  
E. Skovlund ◽  
H.G.M. Leufkens ◽  
J.F. Smyth

2020 ◽  
Vol 10 (1) ◽  
pp. 1877-1888

The world of medicine explored the use of nanoparticles in therapeutics in the last two decades. Owing to the advantages nanoparticles offer, they are proving beneficial to overcome many drawbacks faced by small drug molecules. Since the nature, architecture, shape, size, and mechanism of action of nanomedicines totally different from regularly used drugs, it is important to work on the possible toxicity these nanoparticles are causing so that its safety can be ensured. In today’s scenario, a lot of industries and institutes are synthesizing nano drugs, so it is important to check its toxicity and safety evaluation under in vivo and in vitro conditions, as it has come to fore that number of metal and carbon-based nanoparticles, although proving useful further display increased toxicity. Taken into consideration nanoparticle toxicity and safety, the present review discusses the exact working of nanoparticles at the molecular, cellular, and physiological levels and the toxicity associated with it. The present strategies for safety assessment have also been reviewed. The research involving nanomaterials in therapeutics demand strict regulation in nanoparticle synthesis, its usage, properly regulated clinical trials ensuring safety assessment.


Theranostics ◽  
2018 ◽  
Vol 8 (19) ◽  
pp. 5259-5275 ◽  
Author(s):  
Yeonho Jo ◽  
Nakwon Choi ◽  
Kyobum Kim ◽  
Hyung-Jun Koo ◽  
Jonghoon Choi ◽  
...  

2014 ◽  
Vol 70 (1) ◽  
pp. 73-85 ◽  
Author(s):  
Antonello Caruso ◽  
Nicolas Frances ◽  
Christophe Meille ◽  
Andrea Greiter-Wilke ◽  
Alexander Hillebrecht ◽  
...  

2019 ◽  
Vol 43 (17) ◽  
pp. 6622-6635 ◽  
Author(s):  
Sevinc Ilkar Erdagi ◽  
Ufuk Yildiz

In this study, a polymeric nanoparticle-mediated dual anti-cancer drug delivery system was designed and developed.


ADMET & DMPK ◽  
2018 ◽  
Vol 6 (1) ◽  
pp. 4
Author(s):  
Godefridus J. Peters ◽  
Anne-Sophie Govaerts ◽  
Hans R. Hendriks ◽  
For EORTC Pharmacology and Molecular Mechanism Group

<p>Drug development consists of many sequential and parallel steps; failure in one of the steps can lead to discontinuation of the process. The process is time-consuming and very expensive, especially the clinical phase. In order to enhance cancer drug development in the 1980s, the National Cancer Institute (NCI) adopted a new screening system using 60 different tumour cell lines from various histologies.  All standard drugs were tested in this panel and it is still open for testing of new chemical entities (NCE) of potential interest. The European NCI compounds initiative, a collaborative programme of the NCI, the Cancer Research Campaign (CRC; now CRUK) and the Pharmacology and Molecular Mechanism Group (PAMM) of the EORTC (European Organization on Research and Treatment of Cancer), was initiated in 1993. The programme aimed to help the NCI reducing its backlog of in vivo testing by further evaluation of interesting European compounds using a pharmacologically directed approach. Considerable multidisciplinary expertise in drug development was combined by the CRC and EORTC-PAMM: chemists, pharmacists, biologists, pharmacologists, oncologists. Selection criteria for European NCI compounds included novelty of the NCE, in vitro activity, if available in vivo and hollow fibre activity, and COMPARE negativity. Over a period of more than 20 years 95 out of approximately 2,000 reviewed compounds were selected. These compounds were put through a series of stepwise pharmacological tests comprising solubility (suitable formulation to administer the NCE to mice), feasibility to develop a simple analytical assay (usually HPLC), limited toxicology and angiogenic properties. This paper provides examples to illustrate the rigorousness of the elimination process of the compounds and discusses the way to improve the process by inclusion of more physico-chemical parameters.</p>


Sign in / Sign up

Export Citation Format

Share Document