scholarly journals Premature Clinical Trial Discontinuation in the Era of Immune Checkpoint Inhibitors

2018 ◽  
Vol 23 (12) ◽  
pp. 1494-1499 ◽  
Author(s):  
Monica Khunger ◽  
Sagar Rakshit ◽  
Adrian V. Hernandez ◽  
Vinay Pasupuleti ◽  
Kate Glass ◽  
...  
2018 ◽  
Vol 17 (12) ◽  
pp. 854-855 ◽  
Author(s):  
Jun Tang ◽  
Jia Xin Yu ◽  
Vanessa M. Hubbard-Lucey ◽  
Svetoslav T. Neftelinov ◽  
Jeffrey P. Hodge ◽  
...  

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e17112-e17112
Author(s):  
Lydia Glick ◽  
Cassra Clark ◽  
Timothy M. Han ◽  
James Ryan Mark ◽  
Leonard G. Gomella ◽  
...  

e17112 Background: Immune Checkpoint Inhibitors (ICI) are increasingly utilized for genitourinary (GU) malignancies. However, data is lacking on the efficacy of these drugs in “real-world” populations - patients who do not fit the strict clinical trial criteria, but may still benefit from therapy. We performed a retrospective analysis of patients receiving ICI at a single tertiary-care institution, with special attention to clinical trial enrollment, adverse events, progression and survival. Methods: Patients receiving ICI for GU malignancies at Thomas Jefferson University Hospital from January 2017 to January 2019 were identified. Descriptive statistics of treatment and pathologies were performed. Progression-free survival (PFS) was calculated from start of ICI to documentation of progression or last follow-up. PFS and overall survival were assessed using Kaplan Meier log-rank test, stratified by trial enrollment. Results: 111 patients were included: 37 on ICI clinical trial, 70 received ICI “off-trial” and 4 received ICI in both settings. 11 patients (10%) underwent multiple courses of ICI throughout treatment. The number of patients initiating ICI increased annually; by 2018, the number of patients initiated on ICI “off-trial” exceeded those initiating ICI “on-trial” (79% vs 21%). Treated pathology included Urothelial Carcinoma (UC; 42%), Renal Cell Carcinoma (RCC; 28%), and Prostate Adenocarcinoma (PCa; 20%). “Off-trial” ICI was more often administered later in the disease course, compared to a more even distribution of “on-trial” ICI administration. Mean PFS and OS for both cohorts can be seen in Table. Conclusions: As seen in our single-institution referral center, the use of immune checkpoint inhibitors has significantly increased – and is now more commonly used off-trial than on-trial. As their use becomes more common, their efficacy in “off-trial” populations must be further investigated. [Table: see text]


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A703-A703
Author(s):  
Krish Perumal ◽  
Amin Osmani

BackgroundSince the approval of the first immune checkpoint inhibitor (ICI) targeting CTLA-4 in 2011 (ipilimumab), six others, targeting the PD-1/PD-L1, have been approved by FDA for a total of more than 19 indications,6,7,8 and the number is growing. These approvals paved the way for rapid growth in the number of candidates in the pipelines. It is critical for these candidates to pursue the right development strategy to demonstrate their potential to regulatory authorities and reach patients without delay. Unexpected challenges in such a competitive field risks leading to expensive modifications and possible discontinuations. This is compounded by the lack of clarity in important development questions such as study design,5 the choice of endpoints and appropriate statistical methods.1,2,3 In this regard, FDA’s guidance document4 provides a useful summary of the topics encountered by clinical development practitioners such as endpoints, clinical trial design and statistical analysis. However, it does not capture the unique challenges of the checkpoint inhibitor space, namely traditional phase I study designs and their ability to predict dosing and detect dose-related toxicities1 and endpoint selection given the unconventional response patterns.2MethodsThe approval packages of the seven FDA-approved ICIs contain a wealth of information related to the focus areas, expectations and concerns of the agency. However, they run into thousands of pages, which renders manual analysis too time-consuming and/or incomplete. In this work, we use Regulatory Foresight, a proprietary AI software tool developed by Biotech Square Inc., that employs state-of-the-art techniques in Computer Vision, Natural Language Processing and Machine Learning to extract, standardize, and analyze interactions from drug and biologic applications reviewed by FDA.ResultsUsing Regulatory Foresight, we discovered (a) the major topics of interest and concerns of the FDA, (b) the commonalities and differences in topics between the individual ICIs, (c) the evolution of topics from the oldest to the most recently approved ICI, and (d) the unaddressed topics in official FDA guidance documents.ConclusionsThis work successfully uncovers regulatory requirements in the development of immune checkpoint inhibitors using AI algorithms in order for sponsors to (a) optimize strategies for development of new drugs, (b) better understand regulatory expectations, and (c) adequately prepare for meetings and submissions to regulatory agencies. In addition this work discovers the current gaps in official FDA guidance documents so that they may be adequately addressed in future versions.ReferencesJardim DL, de Melo Gagliato D, Kurzrock R. Lessons From the Development of the Immune Checkpoint Inhibitors in Oncology. Integr Cancer Ther 2018;17(4):1012–1015. doi:10.1177/1534735418801524Ferrara R, Pilotto S, Caccese M, et al. Do immune checkpoint inhibitors need new studies methodology?. J Thorac Dis2018;10(Suppl 13):S1564–S1580. doi:10.21037/jtd.2018.01.131‘Impact Story: Determining the Clinical Benefit of Treatment Beyond Progression with Immune Checkpoint Inhibitors’ US FDA Regulatory Science Impact Story. Accessed on the 26th of July 2020: https://www.fda.gov/drugs/regulatory-science-action/impact-story-determining-clinical-benefit-treatment-beyond-progression-immune-checkpoint-inhibitors‘Clinical trial endpoints for the approval of cancer drugs and biologics’ Guidance for industry. US Department of Health and Human Services, FDA, Oncology Center of Excellence, CDER, CBER. December 2018Gong, J., Chehrazi-Raffle, A., Reddi, S. et al. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J. Immunotherapy cancer 2018;6:8. https://doi.org/10.1186/s40425-018-0316-zVaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of Indications of FDA-Approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers (Basel) 2020;12(3):738. Published 2020 Mar 20. doi:10.3390/cancers12030738PD1/PD-L1Landscape. Cancer Research Institute. Accessed on the 26th of July 2020. https://www.cancerresearch.org/scientists/immuno-oncology-landscape/pd-1-pd-l1-landscapeImmuno-Oncology Landscape. Cancer Research Institute. Accessed on the 26th of July 2020. https://www.cancerresearch.org/scientists/immuno-oncology-landscape


2020 ◽  
Vol 8 (2) ◽  
pp. e000970
Author(s):  
Kyoung-Ho Pyo ◽  
Sun Min Lim ◽  
Chae-Won Park ◽  
Ha-Ni Jo ◽  
Jae Hwan Kim ◽  
...  

BackgroundEML4-ALK is a distinct molecular entity that is highly sensitive to ALK tyrosine kinase inhibitors (TKIs). Immune checkpoint inhibitors (ICIs) have not proved efficacy in ALK-positive non-small cell lung cancer so far. In this study, we performed a mouse clinical trial using EML4-ALK transgenic mice model to comprehensively investigate immunomodulatory effects of ALK TKI and to investigate the mechanisms of resistance to ICIs.MethodsEML4-ALK transgenic mice were randomized to three treatment arms (arm A: antiprogrammed death cell protein-1 (PD-1), arm B: ceritinib, arm C: anti-PD-1 and ceritinib), and tumor response was evaluated using MRI. Progression-free survival and overall survival were measured to compare the efficacy. Flow cytometry, multispectral imaging, whole exome sequencing and RNA sequencing were performed from tumors obtained before and after drug resistance.ResultsMouse clinical trial revealed that anti-PD-1 therapy was ineffective, and the efficacy of ceritinib and anti-PD-1 combination was not more effective than ceritinib alone in the first line. Dynamic changes in immune cells and cytokines were observed following each treatment, while changes in T lymphocytes were not prominent. A closer look at the tumor immune microenvironment before and after ceritinib resistance revealed increased regulatory T cells and programmed death-ligand 1 (PD-L1)-expressing cells both in the tumor and the stroma. Despite the increase of PD-L1 expression, these findings were not accompanied by increased effector T cells which mediate antitumor immune responses.ConclusionsALK-positive tumors progressing on ceritinib is not immunogenic enough to respond to immune checkpoint inhibitors.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 11531-11531
Author(s):  
Hazel O'Sullivan ◽  
Dearbhaile Collins ◽  
Deirdre O'Mahony ◽  
Derek Power ◽  
Richard Bambury ◽  
...  

11531 Background: In the era of cytotoxic chemotherapy, aggressive cancer treatment and hospitalization at the end of life (EOL) has been associated with a worse quality of death. Meanwhile, in the era of immunotherapy (IO), little is known of the impact of these novel agents on EOL care. The aim of this study was to evaluate the EOL care of metastatic cancer patients treated with immune checkpoint inhibitors. Methods: We conducted a retrospective analysis of patients prescribedPD1/L1 or CTLA-4 antibodies in Cork University Hospital (CUH) and Mercy University Hospital (MUH) between January 2013 to December 2018. Patients treated on a clinical trial were excluded. Results: We identified 224 patients treated with immune checkpoint inhibitors (outside of a clinical trial) in CUH and MUH over the described 6 year period. 108 of these patients were deceased, 102 electronic files were available for analysis. Of the 102 patients, 57 had metastatic melanoma, 33 non small cell lung cancer, 8 renal cell carcinoma, 4 had other advanced malignancies. 43% were female and 57% were male. 6% of patients had an ECOG performance status (PS) of 0 at diagnosis, 80% PS of 1 and 10% PS of 2. Median age at death was 62 years. 47 patients were treated with pembrolizumab, 26 nivolumab, 25 ipilimumab, 2 nivolumab/ipilimumab and 2 received atezolizumab. 29 patients received IO as first line treatment, 50 as second line, 17 as third line and 6 as fourth line. Median number of IO cycles received was 4 (range 1 - 41). Progression of disease (62%) and declining performance status (14%) were the most common reasons for discontinuation of IO treatment. 16 of the 102 patients received a further line of systemic therapy. Median time from last dose of IO to death was 57 days. 20 patients (20%) died within 30 days of last dose of IO. Of these 20 patients, the median number of cycles of IO received was 2 (range 1-7), 8 of these 20 patients received one cycle of IO only. 39 patients (38%) attended the ED in the last month of life. 47 (46%) patients had at least one hospital admission in the last month of life, the median hospital length of stay was 6 days (range 1-30) and 22 patients died in hospital. 94% of patients were referred to palliative care, the median time from palliative care referral to death was 64 days (range 1- 1010), 62% of patients died in hospice. Conclusions: Patients with advanced cancer treated with immunotherapy have high rates of hospital admissions and ED attendances despite early palliative care involvement. 20% of patients died within 30 days of IO. More research is needed to help physicians identify patients who are least likely to benefit from IO so as not to treat futile cases.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS11579-TPS11579
Author(s):  
Yasuhiro Fujisawa ◽  
Koji Yoshino ◽  
Taiki Isei ◽  
Kenjiro Namikawa ◽  
Taku Fujimura ◽  
...  

TPS11579 Background: Cutaneous angiosarcoma (CAS) is a rare sarcoma and advanced cases face a complex treatment regimen and dismal prognosis. Several cytotoxic agents (anthracycline, taxane, and gemcitabine) and targeted therapies (bevacizumab and sorafenib) have been tested for advanced disease in clinical trials with poor results. Since CAS commonly develops in elderly patients, high-grade adverse events from treatment may not be ideal and thus therapies featuring durable response and low comorbidity are in great demand. Recent studies suggest that CAS has a distinct genomic profile, including higher tumor mutational burden (TMB), compared to angiosarcomas developed in other sites. Moreover, we have shown that CAS with higher PD-1/L1 in the tumor microenvironment had statistically better overall survival compared with those without, indicating CAS susceptibility to immune checkpoint blockade therapy. However, to date, reports of immune checkpoint inhibitors for CAS are nonexistent. Methods: The present study is a phase 2, multicenter, single-arm clinical trial of ONO-4538 (nivolumab, 480 mg IV every 4 weeks) for patients with unresectable or metastatic CAS refractory to first-line paclitaxel. Twenty-three patients with advanced CAS will be enrolled at 11 sites in Japan with a primary objective to assess the confirmed response rate (H0 p < 5%, H1: p > 20%) to nivolumab. Secondary endpoints include PFS, OS, time to response, and adverse events. A correlative aim includes assessing tissue biomarkers using whole-genome sequencing (1023 genes including interferon-gamma associated genes and other known factors associated with response to immune checkpoint inhibitors) for association with response to nivolumab. The study started in April 2020 and remains open with 7 patients enrolled at time of Clinical trial information: UMIN000043039.


Sign in / Sign up

Export Citation Format

Share Document