proof of mechanism
Recently Published Documents


TOTAL DOCUMENTS

58
(FIVE YEARS 17)

H-INDEX

15
(FIVE YEARS 2)

BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Stephan Ursprung ◽  
Helen Mossop ◽  
Ferdia A. Gallagher ◽  
Evis Sala ◽  
Richard Skells ◽  
...  

Abstract Background Window-of-opportunity trials, evaluating the engagement of drugs with their biological target in the time period between diagnosis and standard-of-care treatment, can help prioritise promising new systemic treatments for later-phase clinical trials. Renal cell carcinoma (RCC), the 7th commonest solid cancer in the UK, exhibits targets for multiple new systemic anti-cancer agents including DNA damage response inhibitors, agents targeting vascular pathways and immune checkpoint inhibitors. Here we present the trial protocol for the WIndow-of-opportunity clinical trial platform for evaluation of novel treatment strategies in REnal cell cancer (WIRE). Methods WIRE is a Phase II, multi-arm, multi-centre, non-randomised, proof-of-mechanism (single and combination investigational medicinal product [IMP]), platform trial using a Bayesian adaptive design. The Bayesian adaptive design leverages outcome information from initial participants during pre-specified interim analyses to determine and minimise the number of participants required to demonstrate efficacy or futility. Patients with biopsy-proven, surgically resectable, cT1b+, cN0–1, cM0–1 clear cell RCC and no contraindications to the IMPs are eligible to participate. Participants undergo diagnostic staging CT and renal mass biopsy followed by treatment in one of the treatment arms for at least 14 days. Initially, the trial includes five treatment arms with cediranib, cediranib + olaparib, olaparib, durvalumab and durvalumab + olaparib. Participants undergo a multiparametric MRI before and after treatment. Vascularised and de-vascularised tissue is collected at surgery. A ≥ 30% increase in CD8+ T-cells on immunohistochemistry between the screening and nephrectomy is the primary endpoint for durvalumab-containing arms. Meanwhile, a reduction in tumour vascular permeability measured by Ktrans on dynamic contrast-enhanced MRI by ≥30% is the primary endpoint for other arms. Secondary outcomes include adverse events and tumour size change. Exploratory outcomes include biomarkers of drug mechanism and treatment effects in blood, urine, tissue and imaging. Discussion WIRE is the first trial using a window-of-opportunity design to demonstrate pharmacological activity of novel single and combination treatments in RCC in the pre-surgical space. It will provide rationale for prioritising promising treatments for later phase trials and support the development of new biomarkers of treatment effect with its extensive translational agenda. Trial registration ClinicalTrials.gov: NCT03741426 / EudraCT: 2018–003056-21.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 2653-2653
Author(s):  
Abdulazeez Salawu ◽  
Rachel Chen ◽  
Alberto Hernando-Calvo ◽  
Daniel Vilarim Araujo ◽  
Marc Oliva Bernal ◽  
...  

2653 Background: Pharmacodynamic biomarkers (PD) are considered fundamental for go/no-go decisions in phase 1 trials. Despite an increase in the availability of blood-based biomarker assays, the requirement of invasive non-diagnostic research tumor biopsies for trial eligibility remains common. In the immuno-oncology (IO) era, the impact of PD analysis for the confirmation of biologic activity and recommended phase 2 dose (RP2D) has not been investigated. Methods: Phase 1 studies from 01/2014 to 12/2018 were reviewed. Among 12053 abstracts screened, a total of 143 phase I-IO trials were identified. Characteristics of studies that included on-treatment PD biomarkers (tissue-derived, blood-based and radiomic) were extracted and analyzed. Outcomes from the biomarker data in terms of proof of mechanism/biologic activity and statistically significant correlation with clinical benefit (objective response or survival) were collected. Authors’ statements on the influence of PD results on RP2D were also noted. Results: Out of 143 phase 1 IO trials, 107 (75%) were monotherapy. The most frequent IO evaluated were vaccines (41%), cell therapy (16%), immunomodulators (13%) and cytokines (7%). Of the 36 combination studies, 20 (61%) included a second IO drug while 16 (39%) included molecular-targeted agents. Only 18 of 143 studies (12%) did not report any PD data. Of the remaining 125 studies, tissue-derived PD (t-PD) biomarkers alone, blood-based PD (b-PD) biomarkers alone, both t-PD and b-PD biomarkers, and imaging biomarkers were tested in 3 (2%), 97 (78%), 25 (20%), and 7 (6%), respectively. Demonstration of proof of mechanism/biologic activity only were reported in 16/28 (57%), 80/122 (66%) and 4/7 (57%) of the t-PD, b-PD and imaging biomarker studies, respectively. Significant correlation with clinical benefit was reported in 2/28 (7%), 7/122 (6%) and 0/7 (0%) of the t-PD, b-PD and imaging biomarker studies, respectively; these involved 4 vaccines (1 in combination with PD1 blockade), 1 cell therapy and 1 oncolytic virus (in combination with CTLA4 blockade). Among 35 b-PD studies with negative results, 5 also performed t-PD biomarkers, all with negative results. Notably, 3 out of 10 t-PD studies with negative results reported concurrent positive b-PD results. Based on the published reports, authors stated that biomarker results helped with RP2D determination in 16/28 (57%) of t-PD and 78/122 (64%) of b-PD studies. Conclusions: Our results suggest that in the IO era, most studies perform PD analysis, with similar proportions of t-PD and b-PD showing proof of mechanism/biologic activity. IO PD biomarkers have limited correlation with clinical benefit. Many authors considered IO PD biomarkers to be relevant in RP2D decisions, but this needs confirmation by other measures of impact. With continued technological developments utilizing circulating biomarkers, b-PD may ultimately replace many t-PD tests in future IO studies.


Author(s):  
Daniel Umbricht ◽  
Markus Abt ◽  
Paul Tamburri ◽  
Christopher Chatham ◽  
Štefan Holiga ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A6-A6
Author(s):  
Russell Garland ◽  
Christopher Kirkham ◽  
Michelle Yap ◽  
Louise Brackenbury ◽  
Tommaso Iannitti ◽  
...  

BackgroundLack of efficacy is a common cause of failure in Phase I and Phase II clinical trials. Pharmacodynamic (PD) biomarker assays can demonstrate target engagement and proof of mechanism; both key components to improve trial success. Biomarkers established at the pre-clinical phase can serve as exploratory endpoints in early phase clinical trials, to confirm the mode of action of the therapeutic. We show examples of human in vitro assays and murine T cell adoptive transfer models, which can be used to establish potential PD biomarkers for inclusion in the clinical phases.MethodsHuman peripheral blood mononuclear cell (PBMC) were incubated with SEB in the presence of Pembrolizumab or Ipilimumab. IL-2 and IFNgamma levels were quantified by Luminex. To identify biomarkers of checkpoint inhibition, mice transferred with a defined population of ovalbumin (OVA)-specific T cells were challenged with OVA antigen or EG7 tumour. Activation and proliferation of antigen-specific T cells was determined and Nanostring gene expression analysis performed. Flow cytometry staining panels for human immune markers including CD4, CD14, CD25 and FOXP3 were established pre-clinically. As part of the assay validation process for a clinical trial, whole blood SEB activation was performed in normal donors, with Luminex analysis of IL-2, IL-17, IFNgamma and TNFalpha.ResultsImmune checkpoint inhibitors resulted in increased IL-2 and IFNgamma secretion in human PBMC stimulated with SEB. In the murine PD model, anti-PD-L1 caused upregulation of CD25, IFNgamma and granzyme B by antigen-specific CD8 T cells. Gene expression analysis of murine tumours elucidated changes in response to a vaccine. Flow cytometry panel staining determined the frequencies of human Treg and monocytes, which are common targets of immune-modulating therapies. Fit-for-purpose validation was performed for a human SEB activation assay resulting in robust changes in cytokine production.ConclusionsThe experiments here show the flow of experiments that can be performed to identify a PD biomarker for use in first in man trials; the pre-clinical human PBMC SEB screening assay provides a simple assay demonstrating that a therapy can enhance T cell function and would be translatable to the clinic. The murine PD model provides a platform to screen for biomarkers of T cell function and monitor gene expression modulation. Biomarkers identified in the murine setting provide a good starting point for exploratory assessment in early phase clinical trials, where inclusion of exploratory PD biomarker endpoints in can confirm proof of mechanism and improve study success rates.Ethics ApprovalHuman tissues used in this study were collected with ethical approval from UK Research Ethics Committee South West, Bristol (UK), approval number 15/SW/0029.


2020 ◽  
Vol 12 (1) ◽  
Author(s):  
Harald Hampel ◽  
◽  
Simone Lista ◽  
Eugeen Vanmechelen ◽  
Henrik Zetterberg ◽  
...  

Abstract β-Secretase1 (BACE1) protein concentrations and rates of enzyme activity, analyzed in human bodily fluids, are promising candidate biological markers for guidance in clinical trials investigating BACE1 inhibitors to halt or delay the dysregulation of the amyloid-β pathway in Alzheimer’s disease (AD). A robust body of evidence demonstrates an association between cerebrospinal fluid/blood BACE1 biomarkers and core pathophysiological mechanisms of AD, such as brain protein misfolding and aggregration, neurodegeneration, and synaptic dysfunction. In pharmacological trials, BACE1 candidate biomarkers may be applied to a wide set of contexts of use (CoU), including proof of mechanism, dose-finding, response and toxicity dose estimation. For clinical CoU, BACE1 biomarkers show good performance for prognosis and disease prediction. The roadmap toward validation and qualification of BACE1 biomarkers requires standardized pre-analytical and analytical protocols to reduce inter-site variance that may have contributed to inconsistent results. BACE1 biomarker-drug co-development programs, including biomarker-guided outcomes and endpoints, may support the identification of sub-populations with a higher probability to benefit from BACE1 inhibitors with a reduced risk of adverse effects, in line with the evolving precision medicine paradigm.


2020 ◽  
Vol 45 (11) ◽  
pp. 1842-1850 ◽  
Author(s):  
Joshua T. Kantrowitz ◽  
Jack Grinband ◽  
Donald C. Goff ◽  
Adrienne C. Lahti ◽  
Stephen R. Marder ◽  
...  

Abstract Glutamate neurotransmission is a prioritized target for antipsychotic drug development. Two metabotropic glutamate receptor 2/3 (mGluR2/3) agonists (pomaglumetad [POMA] and TS-134) were assessed in two Phase Ib proof of mechanism studies of comparable designs and using identical clinical assessments and pharmacoBOLD methodology. POMA was examined in a randomized controlled trial under double-blind conditions for 10-days at doses of 80 or 320 mg/d POMA versus placebo (1:1:1 ratio). The TS-134 trial was a randomized, single-blind, 6-day study of 20 or 60 mg/d TS-134 versus placebo (5:5:2 ratio). Primary outcomes were ketamine-induced changes in pharmacoBOLD in the dorsal anterior cingulate cortex (dACC) and symptoms reflected on the Brief Psychiatric Rating Scale (BPRS). Both trials were conducted contemporaneously. 95 healthy volunteers were randomized to POMA and 63 to TS-134. High-dose POMA significantly reduced ketamine-induced BPRS total symptoms within and between-groups (p < 0.01, d = −0.41; p = 0.04, d = −0.44, respectively), but neither POMA dose significantly suppressed ketamine-induced dACC pharmacoBOLD. In contrast, low-dose TS-134 led to moderate to large within and between group reductions in both BPRS positive symptoms (p = 0.02, d = −0.36; p = 0.008, d = −0.82, respectively) and dACC pharmacoBOLD (p = 0.004, d = −0.56; p = 0.079, d = −0.50, respectively) using pooled across-study placebo data. High-dose POMA exerted significant effects on clinical symptoms, but not on target engagement, suggesting a higher dose may yet be needed, while the low dose of TS-134 showed evidence of symptom reduction and target engagement. These results support further investigation of mGluR2/3 and other glutamate-targeted treatments for schizophrenia.


2020 ◽  
Vol 26 (5) ◽  
pp. 760-768 ◽  
Author(s):  
Andrew D. Krystal ◽  
Diego A. Pizzagalli ◽  
Moria Smoski ◽  
Sanjay J. Mathew ◽  
John Nurnberger ◽  
...  
Keyword(s):  

Sign in / Sign up

Export Citation Format

Share Document