1
|
Dhillon S, Lopes G, Parker JL. The Effect of Biomarkers on Clinical Trial Risk in Gastric Cancer. Am J Clin Oncol 2023; 46:58-65. [PMID: 36662871 DOI: 10.1097/coc.0000000000000963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVES This study examined clinical trial success rates for new drug developments in gastric cancer since 1998. We also examined the clinical trial design features that may mitigate the risk of clinical trial failure. MATERIALS AND METHODS Clinical trial data was obtained from clinicaltrials.gov. Drugs were included if they entered testing between January 1, 1998 and January 1, 2022 and were excluded if they did not have a completed phase I trial or treated secondary effects of gastric cancer. Transition probabilities were calculated for each phase and compared with industry averages. Success rates were determined based on biomarker usage, drug target, type of therapy, and drug chemistry. RESULTS Upon screening 1990 trials, 219 drugs met our inclusion criteria. The probability of a drug completing all phases of testing and obtaining FDA approval was 7%, which is below the 11% industry average. The use of biomarkers in clinical development resulted in nearly a 2-fold increase in the cumulative success rate. Biologics also exhibited higher success rates (17%) as opposed to small molecules (1%). This was true even when we compared both drug types that shared the same target. When comparing only receptor-targeted therapies, biologics (62%) continued to outperform small molecules (18%). Similarly, when narrowed down to drugs targeting solely HER2 receptors, biologics continued to prevail (64% vs. 24%). CONCLUSIONS Implementing biomarkers, receptor-targeted therapies, and biologics in clinical development improves clinical trial success rates in gastric cancer. Thus, physicians should prioritize the enrollment of gastric cancer patients in clinical trials that incorporate the aforementioned features.
Collapse
Affiliation(s)
- Sumeet Dhillon
- Department of Biology, University of Toronto Mississauga, Mississauga, ON
| | - Gilberto Lopes
- University of Miami, Miller School of Medicine, Miami, FL
| | - Jayson L Parker
- Department of Biology, University of Toronto Mississauga, Mississauga, ON
| |
Collapse
|
2
|
Mohamed L, Manjrekar S, Ng DP, Walsh A, Lopes G, Parker JL. The Effect of Biomarker Use on the Speed and Duration of Clinical Trials for Cancer Drugs. Oncologist 2022; 27:849-856. [PMID: 35993585 PMCID: PMC9526484 DOI: 10.1093/oncolo/oyac130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Background The purpose of this study was to explore the effects biomarkers have on the duration and speed of clinical trials in oncology. Materials and Methods Clinical trial data was pooled from www.clinicaltrials.gov within the 4 cancer indications of non-small cell lung cancer, breast cancer, melanoma, and colorectal cancer. Heatmaps of clinical timelines were used to display differences in the frequency and timing of clinical trials across trials that used or did not use biomarkers, for all 4 indications. Results Screening of 8630 clinical trials across the 4 indications yielded 671 unique drugs corresponding to 1224 eligible trials used in our analysis. The constructed heatmaps visually represented that biomarkers did not have an effect on the time gap between trial phases for non-small cell lung cancer and melanoma but did for colorectal and breast cancer trials, reducing the speed of trial timelines. It was also observed that biomarker trials were more often concurrent over shorter periods of time and began later in the timeline for non-small cell lung and colorectal cancers. Conclusion The novel visualization method revealed longer gaps between trial phases, later clinical trial start times, and shorter periods of concurrently run trials for drugs that used biomarkers. The study highlights that biomarker-driven trials might impact drug approval timelines and need to be considered carefully in clinical development plan.
Collapse
Affiliation(s)
- Luqmaan Mohamed
- Master of Biotechnology Program, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Siddhi Manjrekar
- Master of Biotechnology Program, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Derek P Ng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Alec Walsh
- Cheriton School of Computer Science, University of Waterloo, Waterloo, ON, Canada
| | - Gilberto Lopes
- University of Miami, Miller School of Medicine, Coral Gables, FL, USA
| | - Jayson L Parker
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| |
Collapse
|
3
|
Parker JL, Kuzulugil SS, Pereverzev K, Mac S, Lopes G, Shah Z, Weerasinghe A, Rubinger D, Falconi A, Bener A, Caglayan B, Tangri R, Mitsakakis N. Does biomarker use in oncology improve clinical trial failure risk? A large-scale analysis. Cancer Med 2021; 10:1955-1963. [PMID: 33620160 PMCID: PMC7957156 DOI: 10.1002/cam4.3732] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose To date there has not been an extensive analysis of the outcomes of biomarker use in oncology. Methods Data were pooled across four indications in oncology drawing upon trial outcomes from www.clinicaltrials.gov: breast cancer, non‐small cell lung cancer (NSCLC), melanoma and colorectal cancer from 1998 to 2017. We compared the likelihood drugs would progress through the stages of clinical trial testing to approval based on biomarker status. This was done with multi‐state Markov models, tools that describe the stochastic process in which subjects move among a finite number of states. Results Over 10000 trials were screened, which yielded 745 drugs. The inclusion of biomarker status as a covariate significantly improved the fit of the Markov model in describing the drug trajectories through clinical trial testing stages. Hazard ratios based on the Markov models revealed the likelihood of drug approval with biomarkers having nearly a fivefold increase for all indications combined. A 12, 8 and 7‐fold hazard ratio was observed for breast cancer, melanoma and NSCLC, respectively. Markov models with exploratory biomarkers outperformed Markov models with no biomarkers. Conclusion This is the first systematic statistical evidence that biomarkers clearly increase clinical trial success rates in three different indications in oncology. Also, exploratory biomarkers, long before they are properly validated, appear to improve success rates in oncology. This supports early and aggressive adoption of biomarkers in oncology clinical trials.
Collapse
Affiliation(s)
- Jayson L Parker
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | | | - Kirill Pereverzev
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Stephen Mac
- Institute of Health Policy, Management and Evaluation, University of Toronto, Mississauga, ON, Canada
| | - Gilberto Lopes
- University of Miami, Miller School of Medicine, Coral Gables, FL, USA
| | - Zain Shah
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | | | - Daniel Rubinger
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Adam Falconi
- Department of Pharmacy, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Ayse Bener
- Mechanical and Industrial Engineering, Ryerson University, Toronto, ON, Canada
| | - Bora Caglayan
- Mechanical and Industrial Engineering, Ryerson University, Toronto, ON, Canada
| | - Rohan Tangri
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Nicholas Mitsakakis
- Institute of Health Policy, Management and Evaluation, and Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
De Martini D. Empowering phase II clinical trials to reduce phase III failures. Pharm Stat 2019; 19:178-186. [PMID: 31729173 DOI: 10.1002/pst.1980] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 07/03/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
The large number of failures in phase III clinical trials, which occur at a rate of approximately 45%, is studied herein relative to possible countermeasures. First, the phenomenon of failures is numerically described. Second, the main reasons for failures are reported, together with some generic improvements suggested in the related literature. This study shows how statistics explain, but do not justify, the high failure rate observed. The rate of failures due to a lack of efficacy that are not expected, is considered to be at least 10%. Expanding phase II is the simplest and most intuitive way to reduce phase III failures since it can reduce phase III false negative findings and launches of phase III trials when the treatment is positive but suboptimal. Moreover, phase II enlargement is discussed using an economic profile. As resources for research are often limited, enlarging phase II should be evaluated on a case-by-case basis. Alternative strategies, such as biomarker-based enrichments and adaptive designs, may aid in reducing failures. However, these strategies also have very low application rates with little likelihood of rapid growth.
Collapse
|
5
|
Jørgensen JT, Hersom M. Clinical and Regulatory Aspects of Companion Diagnostic Development in Oncology. Clin Pharmacol Ther 2017; 103:999-1008. [PMID: 29197081 DOI: 10.1002/cpt.955] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/16/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022]
Abstract
Nearly 20 years have passed since the US Food and Drug Administration (FDA) approved the first companion diagnostic and today this type of assay governs the use of 21 different anticancer drugs. The regulators deem these assays essential for the safe and effective use of a corresponding therapeutic product. The companion diagnostic assays are important both during the drug development process as well as essential treatment decision tools after the approval of the drugs.
Collapse
Affiliation(s)
| | - Maria Hersom
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Zhou JJ, Huang Y, Zhang X, Cheng Y, Tang L, Ma X. Eyes absent gene (EYA1) is a pathogenic driver and a therapeutic target for melanoma. Oncotarget 2017; 8:105081-105092. [PMID: 29285235 PMCID: PMC5739622 DOI: 10.18632/oncotarget.21352] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/28/2017] [Indexed: 12/16/2022] Open
Abstract
EYA1 is a DNA repair enzyme that is induced after DNA damage and is upregulated in melanoma. However, its role in pathogenesis and therapeutic targeting of melanoma is unknown. Our objectives are (1) to study the relationship between EYA1 expression levels and melanoma patients’ clinical pathologic parameters including survival; (2) to investigate its impact on cultured melanoma cells in vitro; and (3) to evaluate EYA1 inhibitors’ potential as a treatment of melanoma. Melanoma tissue microarrays were used to assess EYA1 protein expression in 326 melanoma tissues, and to correlate the expression with patients’ clinical pathological parameters. In addition, retroviral ShRNA vectors were used to silence expression of EYA1 in A375 melanoma cells, and the resultant cells examined for changes in growth, DNA synthesis, and tumor formation in vitro. Lastly, melanoma cells were treated with benzbromarone with or without the BRAF inhibitor vemurafenib. Our results showed that EYA1 protein is low in benign nevi, but is significantly up-regulated in melanoma in situ, and remains high in invasive and metastatic melanoma. In addition, silencing of EYA1 gene expression resulted in decreased proliferation and colony formation. These were associated with decreased cyclin D1 and increased phosphorylated histone protein γH2AX. Finally, treatment with benzbromarone, a specific inhibitor of EYA1, caused significant inhibition of melanoma cell proliferation, and increased sensitivity to the BRAF inhibitor vemurafenib. In conclusion, EYA1 gene is a pathogenic driver in melanoma pathogenesis. Targeting EYA1 may be a valuable strategy for treatment of melanoma.
Collapse
Affiliation(s)
- Joshua Jiawei Zhou
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Welichem Biotech Inc., Burnaby, BC, Canada
| | - Yuanshen Huang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
| | - Xue Zhang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
| | - Yabin Cheng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Liren Tang
- Welichem Biotech Inc., Burnaby, BC, Canada
| | - Xiaodong Ma
- College of Pharmacology, Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
7
|
Molecular Markers and Targeted Therapeutics in Metastatic Tumors of the Spine: Changing the Treatment Paradigms. Spine (Phila Pa 1976) 2016; 41 Suppl 20:S218-S223. [PMID: 27488299 DOI: 10.1097/brs.0000000000001833] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY TYPE A review of the literature. OBJECTIVE The aim of this study was to discuss the evolution of molecular signatures and the history and development of targeted therapeutics in metastatic tumor types affecting the spinal column. SUMMARY OF BACKGROUND DATA Molecular characterization of metastatic spine tumors is expected to usher in a revolution in diagnostic and treatment paradigms. Molecular characterization will provide critical information that can be used for initial diagnosis, prognosticating the ideal treatment strategy, assessment of treatment efficacy, surveillance and monitoring recurrence, and predicting complications, clinical outcome, and overall survival in patients diagnosed with metastatic cancers to the spinal column. METHODS A review of the literature was performed focusing on illustrative examples of the role that molecular-based therapeutics have played in clinical outcomes for patients diagnosed with metastatic tumor types affecting the spinal column. RESULTS The impact of molecular therapeutics including receptor tyrosine kinases and immune checkpoint inhibitors and the ability of molecular signatures to provide prognostic information are discussed in metastatic breast cancer, lung cancer, prostate cancer, melanoma, and renal cell cancer affecting the spinal column. CONCLUSION For the providers who will ultimately counsel patients diagnosed with metastases to the spinal column, molecular advancements will radically alter the management/surgical paradigms utilized. Ultimately, the translation of these molecular advancements into routine clinical care will greatly improve the quality and quantity of life for patients diagnosed with spinal malignancies and provide better overall outcomes and counseling for treating physicians. LEVEL OF EVIDENCE N/A.
Collapse
|
8
|
Jørgensen JT. The importance of predictive biomarkers in oncology drug development. Expert Rev Mol Diagn 2016; 16:807-9. [PMID: 27282189 DOI: 10.1080/14737159.2016.1199962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
9
|
Tang R, Xu X, Yang W, Yu W, Hou S, Xuan Y, Tang Z, Zhao S, Chen Y, Xiao X, Huang W, Guo W, Li M, Deng W. MED27 promotes melanoma growth by targeting AKT/MAPK and NF-κB/iNOS signaling pathways. Cancer Lett 2016; 373:77-87. [PMID: 26797421 DOI: 10.1016/j.canlet.2016.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 01/07/2023]
Abstract
The inhibitors of BRAF and MEK targeting MAPK signaling pathway provide a comparatively effective therapeutic strategy for melanoma caused by BRAF mutation. However, melanoma, especially metastatic melanoma, has become one of the most threatening malignancies. Thus, the identification of exact molecular mechanisms and the key components involved in such mechanisms is urgently needed in order to provide new therapeutic options for patients with melanoma. Here, we identified MED27 as a potential melanoma target and explored its role and the associated molecular mechanism involved in melanoma progression. MED27 was found to be highly expressed in melanoma cells and tumor tissues. Its silencing led to melanoma cell proliferation inhibition, cell cycle arrest and apoptosis induction accompanied by the inactivation of PI3K/AKT and MAPK/ERK signaling and the activation of Bax/Cyto-C/Caspase-dependent apoptotic pathway. In addition, silencing of MED27 led to the decrease of iNOS expression through inhibiting the activation of a serial of upstream key proteins of NF-κB signaling pathway and the translocation of p50/p65 from cytoplasm to nucleus. MED27 was also found to be able to interact with NF-κB and p300 and to be acetylated by p300. Furthermore, the results in a xenograft tumor model indicated that melanoma progression was effectively suppressed by MED27 knockdown accompanied by the down-regulation of p-AKT, p-ERK, p-MEK1/2, MMP-9, Bcl-2 and iNOS expressions in the tumor tissues. Taken together, our study not only demonstrated the new function of MED27 as an oncogenic protein and the associated molecular mechanisms involved in melanoma progression, but also provided a possibility for the development of MED27 as a new anticancer target in melanoma therapy.
Collapse
Affiliation(s)
- Ranran Tang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xiangdong Xu
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjing Yang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shuai Hou
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yang Xuan
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhipeng Tang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shilei Zhao
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yiming Chen
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiangsheng Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenlin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| | - Wei Guo
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Man Li
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China.
| |
Collapse
|
10
|
Waldman SA, Terzic A. Companion diagnostics at the intersection of personalized medicine and healthcare delivery. Biomark Med 2015; 9:1-3. [PMID: 25605449 DOI: 10.2217/bmm.14.99] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Scott A Waldman
- Delaware Valley Institute for Clinical & Translational Science, Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, 132 South 10th Street, 1170 Main, Philadelphia, PA 19107, USA
| | | |
Collapse
|
11
|
Hussain HT, Parker JL, Sharma AM. Clinical trial success rates of anti-obesity agents: the importance of combination therapies. Obes Rev 2015. [PMID: 26222385 DOI: 10.1111/obr.12299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The objective of this study was to construct a clinical trial profile assessing the risk of drug failure among anti-obesity agents. Research was conducted by looking at anti-obesity therapies currently on the market or in clinical trials (phases I to III) conducted from 1998 to September 2014, with the exclusion of any drugs whose phase I trial was conducted prior to January 1998. This was completed primarily through a search on http://clinicaltrials.gov where a total of 51 drugs met the search criteria. The transition probabilities were then calculated based on various classifications and compared against industry standards. The transition probability of anti-obesity agents was 8.50% whereas the transition probability of industry standards was 10.40%. Combination therapies had four times the transition probability than monotherapies, 40% and 4.75%, respectively. Therefore, it was determined that 92% of drugs fail during clinical trial testing for this indication and combination therapy appears to improve clinical trial success rates to 10-fold.
Collapse
Affiliation(s)
- H T Hussain
- Biology Department, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - J L Parker
- Biology Department, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - A M Sharma
- Department of Medicine, Division of Endocrinology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|