1
|
Guadagni S, Masedu F, Fiorentini G, Sarti D, Fiorentini C, Guadagni V, Apostolou P, Papasotiriou I, Parsonidis P, Valenti M, Ricevuto E, Bruera G, Farina AR, Mackay AR, Clementi M. Circulating tumour cell gene expression and chemosensitivity analyses: predictive accuracy for response to multidisciplinary treatment of patients with unresectable refractory recurrent rectal cancer or unresectable refractory colorectal cancer liver metastases. BMC Cancer 2022; 22:660. [PMID: 35710393 PMCID: PMC9202660 DOI: 10.1186/s12885-022-09770-3] [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: 02/07/2022] [Accepted: 06/08/2022] [Indexed: 01/19/2023] Open
Abstract
Background Patients with unresectable recurrent rectal cancer (RRC) or colorectal cancer (CRC) with liver metastases, refractory to at least two lines of traditional systemic therapy, may receive third line intraarterial chemotherapy (IC) and targeted therapy (TT) using drugs selected by chemosensitivity and tumor gene expression analyses of liquid biopsy-derived circulating tumor cells (CTCs). Methods In this retrospective study, 36 patients with refractory unresectable RRC or refractory unresectable CRC liver metastases were submitted for IC and TT with agents selected by precision oncotherapy chemosensitivity assays performed on liquid biopsy-derived CTCs, transiently cultured in vitro, and by tumor gene expression in the same CTC population, as a ratio to tumor gene expression in peripheral mononuclear blood cells (PMBCs) from the same individual. The endpoint was to evaluate the predictive accuracy of a specific liquid biopsy precision oncotherapy CTC purification and in vitro culture methodology for a positive RECIST 1.1 response to the therapy selected. Results Our analyses resulted in evaluations of 94.12% (95% CI 0.71–0.99) for sensitivity, 5.26% (95% CI 0.01–0.26) for specificity, a predictive value of 47.06% (95% CI 0.29–0.65) for a positive response, a predictive value of 50% (95% CI 0.01–0.98) for a negative response, with an overall calculated predictive accuracy of 47.22% (95% CI 0.30–0.64). Conclusions This is the first reported estimation of predictive accuracy derived from combining chemosensitivity and tumor gene expression analyses on liquid biopsy-derived CTCs, transiently cultured in vitro which, despite limitations, represents a baseline and benchmark which we envisage will be improve upon by methodological and technological advances and future clinical trials.
Collapse
Affiliation(s)
- Stefano Guadagni
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy.
| | - Francesco Masedu
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Giammaria Fiorentini
- Department of Oncology and Hematology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Pesaro, Italy
| | - Donatella Sarti
- Department of Oncology and Hematology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Pesaro, Italy
| | - Caterina Fiorentini
- Department of Prevention and Sports Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Veronica Guadagni
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | | | | | - Marco Valenti
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Enrico Ricevuto
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Gemma Bruera
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Antonietta R Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Andrew R Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Marco Clementi
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| |
Collapse
|
2
|
Altan M, Kidwell KM, Pelekanou V, Carvajal-Hausdorf DE, Schalper KA, Toki MI, Thomas DG, Sabel MS, Hayes DF, Rimm DL. Association of B7-H4, PD-L1, and tumor infiltrating lymphocytes with outcomes in breast cancer. NPJ Breast Cancer 2018; 4:40. [PMID: 30564631 PMCID: PMC6288133 DOI: 10.1038/s41523-018-0095-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023] Open
Abstract
B7-H4 (VTCN1) is a member of the CD28/B7 family of immune co-inhibitory molecules. The relationship of tumor and stromal B7-H4 protein expression with PD-L1, tumor infiltrating lymphocytes (TILs) and its association with clinico-pathological variables are not well defined. Herein, we explore the expression level of B7-H4 protein in breast cancer and evaluate its association with TILs, levels of PD-L1 expression, and clinico-pathological characteristics in two independent populations. In this study, we used multiplexed automated quantitative immunofluorescence (QIF) to measure the levels of B7-H4 and PD-L1 protein and determined TILs through pathologist assessment of H&E-stained preparations in over a thousand breast cancer cases from two institutions represented in tissue microarray format. Associations between the marker levels, major clinico-pathological variables, and survival were analyzed. We detected B7-H4 protein was highly expressed in both breast cancer and stromal cells. Its expression was independent of breast cancer intrinsic subtypes. PD-L1 expression was higher in triple negative breast cancers. Neither B7-H4 nor PD-L1 were associated with survival in breast cancer. Our study shows there is a mutually exclusive pattern of B7-H4 with both tumor PD-L1 expression and TILs in all breast cancers, independent of breast cancer intrinsic subtype. This exclusive pattern suggests that some breast tumors may preferentially use one B7-related immune evasion mechanism/pathway. This could explain the clinical benefit that is seen only in a fraction of patients with immune checkpoint inhibitors directed exclusively towards PD-L1 in breast cancer.
Collapse
Affiliation(s)
- Mehmet Altan
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Kelley M. Kidwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI USA
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | | | - Daniel E. Carvajal-Hausdorf
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
- Anatomic Pathology, Clinica Alemana-Facultad de Medicina Universidad de Desarrollo, Vitacura, Santiago Chile
| | - Kurt A. Schalper
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Maria I. Toki
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Dafydd G. Thomas
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Michael S. Sabel
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Daniel F. Hayes
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - David L. Rimm
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|
3
|
Abstract
Approximately 75% of patients with pancreatic ductal adenocarcinoma are diagnosed with advanced cancer, which cannot be safely resected. The most commonly used biomarker CA19-9 has inadequate sensitivity and specificity for early detection, which we define as Stage I/II cancers. Therefore, progress in next-generation biomarkers is greatly needed. Recent reports have validated a number of biomarkers, including combination assays of proteins and DNA mutations; however, the history of translating promising biomarkers to clinical utility suggests that several major hurdles require careful consideration by the medical community. The first set of challenges involves nominating and verifying biomarkers. Candidate biomarkers need to discriminate disease from benign controls with high sensitivity and specificity for an intended use, which we describe as a two-tiered strategy of identifying and screening high-risk patients. Community-wide efforts to share samples, data, and analysis methods have been beneficial and progress meeting this challenge has been achieved. The second set of challenges is assay optimization and validating biomarkers. After initial candidate validation, assays need to be refined into accurate, cost-effective, highly reproducible, and multiplexed targeted panels and then validated in large cohorts. To move the most promising candidates forward, ideally, biomarker panels, head-to-head comparisons, meta-analysis, and assessment in independent data sets might mitigate risk of failure. Much more investment is needed to overcome these challenges. The third challenge is achieving clinical translation. To moonshot an early detection test to the clinic requires a large clinical trial and organizational, regulatory, and entrepreneurial know-how. Additional factors, such as imaging technologies, will likely need to improve concomitant with molecular biomarker development. The magnitude of the clinical translational challenge is uncertain, but interdisciplinary cooperation within the PDAC community is poised to confront it.
Collapse
|
4
|
Day RS. Planning clinically relevant biomarker validation studies using the "number needed to treat" concept. J Transl Med 2016; 14:117. [PMID: 27146704 PMCID: PMC4857295 DOI: 10.1186/s12967-016-0862-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/12/2016] [Indexed: 11/21/2022] Open
Abstract
Purpose Despite an explosion of translational research to exploit biomarkers in diagnosis, prediction and prognosis, the impact of biomarkers on clinical practice has been limited. The elusiveness of clinical utility may partly originate when validation studies are planned, from a failure to articulate precisely how the biomarker, if successful, will improve clinical decision-making for patients. Clarifying what performance would suffice if the test is to improve medical care makes it possible to design meaningful validation studies. But methods for tackling this part of validation study design are undeveloped, because it demands uncomfortable judgments about the relative values of good and bad outcomes resulting from a medical decision. Methods An unconventional use of “number needed to treat” (NNT) can structure communication for the trial design team, to elicit purely value-based outcome tradeoffs, conveyed as the endpoints of an NNT “discomfort range”. The study biostatistician can convert the endpoints into desired predictive values, providing criteria for designing a prospective validation study. Next, a novel “contra-Bayes” theorem converts those predictive values into target sensitivity and specificity criteria, to guide design of a retrospective validation study. Several examples demonstrate the approach. Conclusion In practice, NNT-guided dialogues have contributed to validation study planning by tying it closely to specific patient-oriented translational goals. The ultimate payoff comes when the report of the completed study includes motivation in the form of a biomarker test framework directly reflecting the clinical decision challenge to be solved. Then readers will understand better what the biomarker test has to offer patients. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0862-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Roger S Day
- Department of Biomedical Informatics, University of Pittsburgh, 5607 Baum Boulevard, Room 532, Pittsburgh, PA, 15206, USA.
| |
Collapse
|
5
|
Lerner SP, Bajorin DF, Dinney CP, Efstathiou JA, Groshen S, Hahn NM, Hansel D, Kwiatkowski D, O’Donnell M, Rosenberg J, Svatek R, Abrams JS, Al-Ahmadie H, Apolo AB, Bellmunt J, Callahan M, Cha EK, Drake C, Jarow J, Kamat A, Kim W, Knowles M, Mann B, Marchionni L, McConkey D, McShane L, Ramirez N, Sharabi A, Sharpe AH, Solit D, Tangen CM, Amiri AT, Van Allen E, West PJ, Witjes JA, Quale DZ. Summary and Recommendations from the National Cancer Institute's Clinical Trials Planning Meeting on Novel Therapeutics for Non-Muscle Invasive Bladder Cancer. Bladder Cancer 2016; 2:165-202. [PMID: 27376138 PMCID: PMC4927845 DOI: 10.3233/blc-160053] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The NCI Bladder Cancer Task Force convened a Clinical Trials Planning Meeting (CTPM) Workshop focused on Novel Therapeutics for Non-Muscle Invasive Bladder Cancer (NMIBC). Meeting attendees included a broad and multi-disciplinary group of clinical and research stakeholders and included leaders from NCI, FDA, National Clinical Trials Network (NCTN), advocacy and the pharmaceutical and biotech industry. The meeting goals and objectives were to: 1) create a collaborative environment in which the greater bladder research community can pursue future optimally designed novel clinical trials focused on the theme of molecular targeted and immune-based therapies in NMIBC; 2) frame the clinical and translational questions that are of highest priority; and 3) develop two clinical trial designs focusing on immunotherapy and molecular targeted therapy. Despite successful development and implementation of large Phase II and Phase III trials in bladder and upper urinary tract cancers, there are no active and accruing trials in the NMIBC space within the NCTN. Disappointingly, there has been only one new FDA approved drug (Valrubicin) in any bladder cancer disease state since 1998. Although genomic-based data for bladder cancer are increasingly available, translating these discoveries into practice changing treatment is still to come. Recently, major efforts in defining the genomic characteristics of NMIBC have been achieved. Aligned with these data is the growing number of targeted therapy agents approved and/or in development in other organ site cancers and the multiple similarities of bladder cancer with molecular subtypes in these other cancers. Additionally, although bladder cancer is one of the more immunogenic tumors, some tumors have the ability to attenuate or eliminate host immune responses. Two trial concepts emerged from the meeting including a window of opportunity trial (Phase 0) testing an FGFR3 inhibitor and a second multi-arm multi-stage trial testing combinations of BCG or radiotherapy and immunomodulatory agents in patients who recur after induction BCG (BCG failure).
Collapse
Affiliation(s)
| | - Dean F. Bajorin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Colin P. Dinney
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Susan Groshen
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Noah M. Hahn
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Donna Hansel
- University of California, La Jolla, San Diego, CA, USA
| | - David Kwiatkowski
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jonathan Rosenberg
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Robert Svatek
- UT Health Science Center San Antonio, San Antonio, TX, USA
| | - Jeffrey S. Abrams
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Margaret Callahan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Eugene K. Cha
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles Drake
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Jonathan Jarow
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Ashish Kamat
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Kim
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Margaret Knowles
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Bhupinder Mann
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luigi Marchionni
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - David McConkey
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisa McShane
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Nilsa Ramirez
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Andrew Sharabi
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Arlene H. Sharpe
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - David Solit
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Catherine M. Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Eliezer Van Allen
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | | - J. A. Witjes
- Department of Urology, Radboud UMC, Nijmegen, The Netherlands
| | | |
Collapse
|
6
|
Janes H, Brown MD, Pepe MS. Designing a study to evaluate the benefit of a biomarker for selecting patient treatment. Stat Med 2015; 34:3503-15. [PMID: 26112650 PMCID: PMC4626364 DOI: 10.1002/sim.6564] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 03/20/2015] [Accepted: 05/26/2015] [Indexed: 12/31/2022]
Abstract
Biomarkers that predict the efficacy of treatment can potentially improve clinical outcomes and decrease medical costs by allowing treatment to be provided only to those most likely to benefit. We consider the design of a randomized clinical trial in which one objective is to evaluate a treatment selection marker. The marker may be measured prospectively or retrospectively using samples collected at baseline. We describe and contrast criteria around which the trial can be designed. An existing approach focuses on determining if there is a statistical interaction between the marker and treatment. We propose three alternative approaches based on estimating clinically relevant measures of improvement in outcomes with use of the marker. Importantly, our approaches accommodate the common scenario in which the marker-based rule for recommending treatment is developed with data from the trial. Sample sizes are calculated for powering a trial to assess these criteria in the context of adjuvant chemotherapy for the treatment of estrogen-receptor-positive, node-positive breast cancer. In this example, we find that larger sample sizes are generally required for assessing clinical impact than for simply evaluating if there is a statistical interaction between marker and treatment. We also find that retrospectively selecting a case-control subset of subjects for marker evaluation can lead to large efficiency gains, especially if cases and controls are matched on treatment assignment.
Collapse
Affiliation(s)
- Holly Janes
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- University of Washington, Seattle, Washington, USA
| | - Marshall D Brown
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Margaret S Pepe
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Vliegenthart ADB, Antoine DJ, Dear JW. Target biomarker profile for the clinical management of paracetamol overdose. Br J Clin Pharmacol 2015; 80:351-62. [PMID: 26076366 DOI: 10.1111/bcp.12699] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/03/2015] [Accepted: 06/09/2015] [Indexed: 12/21/2022] Open
Abstract
Paracetamol (acetaminophen) overdose is one of the most common causes of acute liver injury in the Western world. To improve patient care and reduce pressure on already stretched health care providers new biomarkers are needed that identify or exclude liver injury soon after an overdose of paracetamol is ingested. This review highlights the current state of paracetamol poisoning management and how novel biomarkers could improve patient care and save healthcare providers money. Based on the widely used concept of defining a target product profile, a target biomarker profile is proposed that identifies desirable and acceptable key properties for a biomarker in development to enable the improved treatment of this patient population. The current biomarker candidates, with improved hepatic specificity and based on the fundamental mechanistic basis of paracetamol-induced liver injury, are reviewed and their performance compared with our target profile.
Collapse
Affiliation(s)
- A D Bastiaan Vliegenthart
- Pharmacology, Toxicology & Therapeutics, University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh
| | - Daniel J Antoine
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - James W Dear
- Pharmacology, Toxicology & Therapeutics, University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh
| |
Collapse
|
8
|
Tian C, Sargent DJ, Krivak TC, Powell MA, Gabrin MJ, Brower SL, Coleman RL. Evaluation of a chemoresponse assay as a predictive marker in the treatment of recurrent ovarian cancer: further analysis of a prospective study. Br J Cancer 2014; 111:843-50. [PMID: 25003664 PMCID: PMC4150278 DOI: 10.1038/bjc.2014.375] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/23/2014] [Accepted: 06/12/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Recently, a prospective study reported improved clinical outcomes for recurrent ovarian cancer patients treated with chemotherapies indicated to be sensitive by a chemoresponse assay, compared with those patients treated with non-sensitive therapies, thereby demonstrating the assay's prognostic properties. Due to cross-drug response over different treatments and possible association of in vitro chemosensitivity of a tumour with its inherent biology, further analysis is required to ascertain whether the assay performs as a predictive marker as well. METHODS Women with persistent or recurrent epithelial ovarian cancer (n=262) were empirically treated with one of 15 therapies, blinded to assay results. Each patient's tumour was assayed for responsiveness to the 15 therapies. The assay's ability to predict progression-free survival (PFS) was assessed by comparing the association when the assayed therapy matches the administered therapy (match) with the association when the assayed therapy is randomly selected, not necessarily matching the administered therapy (mismatch). RESULTS Patients treated with assay-sensitive therapies had improved PFS vs patients treated with non-sensitive therapies, with the assay result for match significantly associated with PFS (hazard ratio (HR)=0.67, 95% confidence interval (CI)=0.50-0.91, P=0.009). On the basis of 3000 simulations, the mean HR for mismatch was 0.81 (95% range=0.66-0.99), with 3.4% of HRs less than 0.67, indicating that HR for match is lower than for mismatch. While 47% of tumours were non-sensitive to all assayed therapies and 9% were sensitive to all, 44% displayed heterogeneity in assay results. Improved outcome was associated with the administration of an assay-sensitive therapy, regardless of homogeneous or heterogeneous assay responses across all of the assayed therapies. CONCLUSIONS These analyses provide supportive evidence that this chemoresponse assay is a predictive marker, demonstrating its ability to discern specific therapies that are likely to be more effective among multiple alternatives.
Collapse
Affiliation(s)
- C Tian
- Precision Therapeutics, Inc., 2516 Jane Street, Pittsburgh, PA 15203, USA
| | - D J Sargent
- Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - T C Krivak
- The Western Pennsylvania Hospital, 4800 Friendship Avenue, Pittsburgh, PA 15224, USA
| | - M A Powell
- Washington University School of Medicine, 4911 Barnes-Jewish Hospital Plaza, St. Louis, MO 63110, USA
| | - M J Gabrin
- Precision Therapeutics, Inc., 2516 Jane Street, Pittsburgh, PA 15203, USA
| | - S L Brower
- Precision Therapeutics, Inc., 2516 Jane Street, Pittsburgh, PA 15203, USA
| | - R L Coleman
- University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Drive, Houston, TX 77030, USA
| |
Collapse
|
9
|
A risk-management approach for effective integration of biomarkers in clinical trials: perspectives of an NCI, NCRI, and EORTC working group. Lancet Oncol 2014; 15:e184-93. [DOI: 10.1016/s1470-2045(13)70607-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
10
|
Dear JW, Antoine DJ. Stratification of paracetamol overdose patients using new toxicity biomarkers: current candidates and future challenges. Expert Rev Clin Pharmacol 2014; 7:181-9. [PMID: 24450481 DOI: 10.1586/17512433.2014.880650] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
One of the most common causes of acute liver failure in the Western world is paracetamol (acetaminophen) overdose. Specific and sensitive detection of liver injury is important for the prompt and safe treatment of patients with the antidote N-acetylcysteine (NAC) and for the determination of NAC efficacy. Despite many years of intense research, the precise mechanisms of paracetamol-induced liver injury in humans are still not defined, and few studies have examined the optimal dosing regimen for clinical NAC use. It has been widely acknowledged that circulating biomarkers such as microRNA-122, keratin-18 and high mobility group box-1 hold potential to inform on the mechanistic-basis of human drug-induced liver injury. Here, we provide a perspective on the application of these mechanistic biomarkers to the deeper understanding of paracetamol hepatotoxicity in clinical and preclinical studies. Also, we discuss current barriers to using these experimental biomarkers to stratify patients presenting to hospital with this common medical emergency.
Collapse
Affiliation(s)
- James W Dear
- National Poisons Information Service Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
11
|
Emmert-Streib F, Abogunrin F, de Matos Simoes R, Duggan B, Ruddock MW, Reid CN, Roddy O, White L, O'Kane HF, O'Rourke D, Anderson NH, Nambirajan T, Williamson KE. Collectives of diagnostic biomarkers identify high-risk subpopulations of hematuria patients: exploiting heterogeneity in large-scale biomarker data. BMC Med 2013; 11:12. [PMID: 23327460 PMCID: PMC3570289 DOI: 10.1186/1741-7015-11-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 01/17/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ineffective risk stratification can delay diagnosis of serious disease in patients with hematuria. We applied a systems biology approach to analyze clinical, demographic and biomarker measurements (n = 29) collected from 157 hematuric patients: 80 urothelial cancer (UC) and 77 controls with confounding pathologies. METHODS On the basis of biomarkers, we conducted agglomerative hierarchical clustering to identify patient and biomarker clusters. We then explored the relationship between the patient clusters and clinical characteristics using Chi-square analyses. We determined classification errors and areas under the receiver operating curve of Random Forest Classifiers (RFC) for patient subpopulations using the biomarker clusters to reduce the dimensionality of the data. RESULTS Agglomerative clustering identified five patient clusters and seven biomarker clusters. Final diagnoses categories were non-randomly distributed across the five patient clusters. In addition, two of the patient clusters were enriched with patients with 'low cancer-risk' characteristics. The biomarkers which contributed to the diagnostic classifiers for these two patient clusters were similar. In contrast, three of the patient clusters were significantly enriched with patients harboring 'high cancer-risk" characteristics including proteinuria, aggressive pathological stage and grade, and malignant cytology. Patients in these three clusters included controls, that is, patients with other serious disease and patients with cancers other than UC. Biomarkers which contributed to the diagnostic classifiers for the largest 'high cancer- risk' cluster were different than those contributing to the classifiers for the 'low cancer-risk' clusters. Biomarkers which contributed to subpopulations that were split according to smoking status, gender and medication were different. CONCLUSIONS The systems biology approach applied in this study allowed the hematuric patients to cluster naturally on the basis of the heterogeneity within their biomarker data, into five distinct risk subpopulations. Our findings highlight an approach with the promise to unlock the potential of biomarkers. This will be especially valuable in the field of diagnostic bladder cancer where biomarkers are urgently required. Clinicians could interpret risk classification scores in the context of clinical parameters at the time of triage. This could reduce cystoscopies and enable priority diagnosis of aggressive diseases, leading to improved patient outcomes at reduced costs.
Collapse
Affiliation(s)
- Frank Emmert-Streib
- Centre for Cancer Research & Cell Biology, Queens University Belfast, Belfast, Northern Ireland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|