1
|
Park Y, Kim MJ, Choi Y, Kim NH, Kim L, Hong SPD, Cho HG, Yu E, Chae YK. Role of mass spectrometry-based serum proteomics signatures in predicting clinical outcomes and toxicity in patients with cancer treated with immunotherapy. J Immunother Cancer 2022; 10:e003566. [PMID: 35347071 PMCID: PMC8961104 DOI: 10.1136/jitc-2021-003566] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2022] [Indexed: 02/03/2023] Open
Abstract
Immunotherapy has fundamentally changed the landscape of cancer treatment. However, only a subset of patients respond to immunotherapy, and a significant portion experience immune-related adverse events (irAEs). In addition, the predictive ability of current biomarkers such as programmed death-ligand 1 (PD-L1) remains unreliable and establishing better potential candidate markers is of great importance in selecting patients who would benefit from immunotherapy. Here, we focus on the role of serum-based proteomic tests in predicting the response and toxicity of immunotherapy. Serum proteomic signatures refer to unique patterns of proteins which are associated with immune response in patients with cancer. These protein signatures are derived from patient serum samples based on mass spectrometry and act as biomarkers to predict response to immunotherapy. Using machine learning algorithms, serum proteomic tests were developed through training data sets from advanced non-small cell lung cancer (Host Immune Classifier, Primary Immune Response) and malignant melanoma patients (PerspectIV test). The tests effectively stratified patients into groups with good and poor treatment outcomes independent of PD-L1 expression. Here, we review current evidence in the published literature on three liquid biopsy tests that use biomarkers derived from proteomics and machine learning for use in immuno-oncology. We discuss how these tests may inform patient prognosis as well as guide treatment decisions and predict irAE of immunotherapy. Thus, mass spectrometry-based serum proteomics signatures play an important role in predicting clinical outcomes and toxicity.
Collapse
Affiliation(s)
- Yeonggyeong Park
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Min Jeong Kim
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yoonhee Choi
- Department of Internal Medicine, NewYork-Presbyterian Queens, Flushing, New York, USA
| | - Na Hyun Kim
- Department of Internal Medicine, AMITA Health Saint Joseph Hospital Chicago, Chicago, Illinois, USA
| | - Leeseul Kim
- Department of Internal Medicine, AMITA Health Saint Francis Hospital Evanston, Evanston, Illinois, USA
| | - Seung Pyo Daniel Hong
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hyung-Gyo Cho
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Emma Yu
- Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Young Kwang Chae
- Department of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
2
|
Semi-Quantitative MALDI Measurements of Blood-Based Samples for Molecular Diagnostics. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030997. [PMID: 35164262 PMCID: PMC8840133 DOI: 10.3390/molecules27030997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/23/2022]
Abstract
Accurate and precise measurement of the relative protein content of blood-based samples using mass spectrometry is challenging due to the large number of circulating proteins and the dynamic range of their abundances. Traditional spectral processing methods often struggle with accurately detecting overlapping peaks that are observed in these samples. In this work, we develop a novel spectral processing algorithm that effectively detects over 1650 peaks with over 3.5 orders of magnitude in intensity in the 3 to 30 kD m/z range. The algorithm utilizes a convolution of the peak shape to enhance peak detection, and accurate peak fitting to provide highly reproducible relative abundance estimates for both isolated peaks and overlapping peaks. We demonstrate a substantial increase in the reproducibility of the measurements of relative protein abundance when comparing this processing method to a traditional processing method for sample sets run on multiple matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) instruments. By utilizing protein set enrichment analysis, we find a sizable increase in the number of features associated with biological processes compared to previously reported results. The new processing method could be very beneficial when developing high-performance molecular diagnostic tests in disease indications.
Collapse
|
3
|
Rich P, Mitchell RB, Schaefer E, Walker PR, Dubay JW, Boyd J, Oubre D, Page R, Khalil M, Sinha S, Boniol S, Halawani H, Santos ES, Brenner W, Orsini JM, Pauli E, Goldberg J, Veatch A, Haut M, Ghabach B, Bidyasar S, Quejada M, Khan W, Huang K, Traylor L, Akerley W. Real-world performance of blood-based proteomic profiling in first-line immunotherapy treatment in advanced stage non-small cell lung cancer. J Immunother Cancer 2021; 9:jitc-2021-002989. [PMID: 34706885 PMCID: PMC8552188 DOI: 10.1136/jitc-2021-002989] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose Immune checkpoint inhibition (ICI) therapy has improved patient outcomes in advanced non-small cell lung cancer (NSCLC), but better biomarkers are needed. A clinically validated, blood-based proteomic test, or host immune classifier (HIC), was assessed for its ability to predict ICI therapy outcomes in this real-world, prospectively designed, observational study. Materials and methods The prospectively designed, observational registry study INSIGHT (Clinical Effectiveness Assessment of VeriStrat® Testing and Validation of Immunotherapy Tests in NSCLC Subjects) (NCT03289780) includes 35 US sites having enrolled over 3570 NSCLC patients at any stage and line of therapy. After enrolment and prior to therapy initiation, all patients are tested and designated HIC-Hot (HIC-H) or HIC-Cold (HIC-C). A prespecified interim analysis was performed after 1-year follow-up with the first 2000 enrolled patients. We report the overall survival (OS) of patients with advanced stage (IIIB and IV) NSCLC treated in the first-line (ICI-containing therapies n=284; all first-line therapies n=877), by treatment type and in HIC-defined subgroups. Results OS for HIC-H patients was longer than OS for HIC-C patients across treatment regimens, including ICI. For patients treated with all ICI regimens, median OS was not reached (95% CI 15.4 to undefined months) for HIC-H (n=196) vs 5.0 months (95% CI 2.9 to 6.4) for HIC-C patients (n=88); HR=0.38 (95% CI 0.27 to 0.53), p<0.0001. For ICI monotherapy, OS was 16.8 vs 2.8 months (HR=0.36 (95% CI 0.22 to 0.58), p<0.0001) and for ICI with chemotherapy OS was unreached vs 6.4 months (HR=0.41 (95% CI 0.26 to 0.67), p=0.0003). HIC results were independent of programmed death ligand 1 (PD-L1). In a subgroup with PD-L1 ≥50% and performance status 0–1, HIC stratified survival significantly for ICI monotherapy but not ICI with chemotherapy. Conclusion Blood-based HIC proteomic testing provides clinically meaningful information for immunotherapy treatment decision in NSCLC independent of PD-L1. The data suggest that HIC-C patients should not be treated with ICI alone regardless of their PD-L1 expression.
Collapse
Affiliation(s)
- Patricia Rich
- Lung Cancer, Piedmont Physicians Group, Atlanta, Georgia, USA
| | | | - Eric Schaefer
- Highlands Oncology Group, Fayetteville, Arkansas, USA
| | - Paul R Walker
- Leo W Jenkins Cancer Center, Brody School of Medicine at East Carolina University, Greenville, North Carolina, USA
| | - John W Dubay
- Lewis and Faye Manderson Cancer Center at DCH Regional Medical Center, Tuscaloosa, Alabama, USA
| | - Jason Boyd
- Southeastern Medical Oncology Center, Goldsboro, North Carolina, USA
| | - David Oubre
- Pontchartrain Cancer Center, Covington, Louisiana, USA
| | - Ray Page
- The Center for Cancer and Blood Disorders, Fort Worth, Texas, USA
| | - Mazen Khalil
- St. Bernards Hospital, Inc, Jonesboro, Arkansas, USA
| | - Suman Sinha
- Christus Saint Michael Health System, Texarkana, Texas, USA
| | - Scott Boniol
- Christus Cancer Treatment Center, Shreveport, Louisiana, USA
| | - Hafez Halawani
- St. Frances Cabrini Hospital Cancer Center, Alexandria, Louisiana, USA
| | - Edgardo S Santos
- Florida Precision Oncology, Division of Genesis Care, Aventura, Florida, USA
| | - Warren Brenner
- Lynn Clinical Research Institute, Boca Raton, Florida, USA
| | | | - Emily Pauli
- Clearview Cancer Institute, Huntsville, Alabama, USA
| | - Jonathan Goldberg
- Clinical Research Alliance, Caremount Medical, Mount Kisco, New York, USA
| | - Andrea Veatch
- Northwest Medical Specialties, Puyallup, Washington, USA
| | - Mitchell Haut
- Hematology and Oncology Associates, Inc, Canton, Ohio, USA
| | | | | | | | | | - Kan Huang
- Phelps County Regional Medical Center, Rolla, Missouri, USA
| | | | - Wallace Akerley
- Huntsman Cancer Institute Cancer Hospital, Salt Lake City, Utah, USA
| |
Collapse
|
4
|
Roder J, Net L, Oliveira C, Meyer K, Asmellash S, Kasimir-Bauer S, Pass H, Weber J, Roder H, Grigorieva J. A proposal for score assignment to characterize biological processes from mass spectral analysis of serum. CLINICAL MASS SPECTROMETRY 2020; 18:13-26. [PMID: 34820522 PMCID: PMC8601010 DOI: 10.1016/j.clinms.2020.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 11/12/2022]
Abstract
Biological process-associated scores generated from mass spectrometry of serum. Scores demonstrated acceptable levels of reproducibility. Scores associated with biological processes and clinical outcome in cancer patients. Possible application to biomarker studies for treatment or monitoring of disease. Multiple biological processes assessed from 3 µL of serum.
Introduction Most diseases involve a complex interplay between multiple biological processes at the cellular, tissue, organ, and systemic levels. Clinical tests and biomarkers based on the measurement of a single or few analytes may not be able to capture the complexity of a patient’s disease. Novel approaches for comprehensively assessing biological processes from easily obtained samples could help in the monitoring, treatment, and understanding of many conditions. Objectives We propose a method of creating scores associated with specific biological processes from mass spectral analysis of serum samples. Methods A score for a process of interest is created by: (i) identifying mass spectral features associated with the process using set enrichment analysis methods, and (ii) combining these features into a score using a principal component analysis-based approach. We investigate the creation of scores using cohorts of patients with non-small cell lung cancer, melanoma, and ovarian cancer. Since the circulating proteome is amenable to the study of immune responses, which play a critical role in cancer development and progression, we focus on functions related to the host response to disease. Results We demonstrate the feasibility of generating scores, their reproducibility, and their associations with clinical outcomes. Once the scores are constructed, only 3 µL of serum is required for the assessment of multiple biological functions from the circulating proteome. Conclusion These mass spectrometry-based scores could be useful for future multivariate biomarker or test development studies for informing treatment, disease monitoring and improving understanding of the roles of various biological functions in multiple disease settings.
Collapse
Key Words
- AIR, acute inflammatory response
- ALK, anaplastic lymphoma kinase
- ANG, angiogenesis
- APR, acute phase reaction
- BRCA1/2, Breast Cancer Gene 1, Breast Cancer Gene 2
- Biological scores
- Biomarker
- CA, complement activation
- CI, confidence interval
- CPH, Cox proportional hazards
- CV, coefficient of variation
- ECM, extracellular matrix organization
- EGFR, epidermal growth factor receptor
- FDA, US Food and Drug Administration
- GLY, glycolysis
- HR, hazard ratio
- HbA1c, hemoglobin A1c
- IFN1, interferon type 1 signaling and response
- IFNg, Interferon γ signaling and response
- IRn, type n immune response
- IT, immune tolerance
- LC MS-MS, liquid chromatography with tandem mass spectrometry
- MALDI ToF, matrix-assisted laser desorption/ionization time of flight
- MRM, multiple reaction monitoring
- MS, mass spectral
- Mass spectrometry
- NSCLC, non-small cell lung cancer
- OS, overall survival
- PC, principal component
- PCA, principal component analysis
- PCn, principal component n
- PD-1, programmed cell death protein 1
- PD-L1, programmed death-ligand 1
- Proteomics
- QC, quality control
- Serum proteome
- Set enrichment analysis
- WH, wound healing
- m/Z, mass/charge
Collapse
Affiliation(s)
- Joanna Roder
- Biodesix, Inc, 2970 Wilderness Place, Boulder, CO 80301, USA
| | - Lelia Net
- Biodesix, Inc, 2970 Wilderness Place, Boulder, CO 80301, USA
| | - Carlos Oliveira
- Biodesix, Inc, 2970 Wilderness Place, Boulder, CO 80301, USA
| | - Krista Meyer
- Biodesix, Inc, 2970 Wilderness Place, Boulder, CO 80301, USA
| | | | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital of Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Harvey Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, 550 1 Ave, New York, NY 10016, USA
| | - Jeffrey Weber
- Perlmutter Cancer Center at NYU Langone Medical Center, 550 1 Ave, New York, NY 10016, USA
| | - Heinrich Roder
- Biodesix, Inc, 2970 Wilderness Place, Boulder, CO 80301, USA
| | | |
Collapse
|
5
|
Definition and Independent Validation of a Proteomic-Classifier in Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12092519. [PMID: 32899818 PMCID: PMC7564837 DOI: 10.3390/cancers12092519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The heterogeneity of epithelial ovarian cancer and its associated molecular biological characteristics are continuously integrated in the development of therapy guidelines. In a next step, future therapy recommendations might also be able to focus on the patient’s systemic status, not only the tumor’s molecular pattern. Therefore, new methods to identify and validate host-related biomarkers need to be established. Using mass spectrometry, we developed and independently validated a blood-based proteomic classifier, stratifying epithelial ovarian cancer patients into good and poor survival groups. We also determined an age dependence of the prognostic performance of this classifier and its association with important biological processes. This work highlights that, just like molecular markers of the tumor itself, the systemic condition of a patient (partly reflected in proteomic patterns) also influences survival and therapy response and could therefore be integrated into future processes of therapy planning. Abstract Mass-spectrometry-based analyses have identified a variety of candidate protein biomarkers that might be crucial for epithelial ovarian cancer (EOC) development and therapy response. Comprehensive validation studies of the biological and clinical implications of proteomics are needed to advance them toward clinical use. Using the Deep MALDI method of mass spectrometry, we developed and independently validated (development cohort: n = 199, validation cohort: n = 135) a blood-based proteomic classifier, stratifying EOC patients into good and poor survival groups. We also determined an age dependency of the prognostic performance of this classifier, and our protein set enrichment analysis showed that the good and poor proteomic phenotypes were associated with, respectively, lower and higher levels of complement activation, inflammatory response, and acute phase reactants. This work highlights that, just like molecular markers of the tumor itself, the systemic condition of a patient (partly reflected in proteomic patterns) also influences survival and therapy response in a subset of ovarian cancer patients and could therefore be integrated into future processes of therapy planning.
Collapse
|
6
|
Nobiletin exerts anti-diabetic and anti-inflammatory effects in an in vitro human model and in vivo murine model of gestational diabetes. Clin Sci (Lond) 2020; 134:571-592. [PMID: 32129440 DOI: 10.1042/cs20191099] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023]
Abstract
Gestational diabetes mellitus (GDM) is a global health issue, whereby pregnant women are afflicted with carbohydrate intolerance with first onset during pregnancy. GDM is characterized by maternal peripheral insulin resistance, thought to be driven by low-grade maternal inflammation. Nobiletin, a polymethoxylated flavonoid, possesses potent glucose-sensitizing and anti-inflammatory properties; however, its effects in GDM have not been assessed. The present study aimed to determine the effects of nobiletin on glucose metabolism and inflammation associated with GDM in both in vitro human tissues and an in vivo animal model of GDM. In vitro, treatment with nobiletin significantly improved TNF-impaired glucose uptake in human skeletal muscle, and suppressed mRNA expression and protein secretion of pro-inflammatory cytokines and chemokines in human placenta and visceral adipose tissue (VAT). Mechanistically, nobiletin significantly inhibited Akt and Erk activation in placenta, and NF-κB activation in VAT. In vivo, GDM mice treated with 50 mg/kg nobiletin daily via oral gavage from gestational day (gd) 1-17 or via i.p. injections from gd 10-17 significantly improved glucose tolerance. Pregnant GDM mice treated with nobiletin from either gd 1-17 or gd 10-17 exhibited significantly suppressed mRNA expression of pro-inflammatory cytokines and chemokines in placenta, VAT and subcutaneous adipose tissue (SAT). Using a quantitative mass spectrometry approach, we identified differentially abundant proteins associated with the effect of nobiletin in vivo. Together, these studies demonstrate that nobiletin improves glucose metabolism and reduces inflammation associated with GDM and may be a novel therapeutic for the prevention of GDM.
Collapse
|
7
|
Muller M, Hummelink K, Hurkmans DP, Niemeijer ALN, Monkhorst K, Roder J, Oliveira C, Roder H, Aerts JG, Smit EF. A Serum Protein Classifier Identifying Patients with Advanced Non-Small Cell Lung Cancer Who Derive Clinical Benefit from Treatment with Immune Checkpoint Inhibitors. Clin Cancer Res 2020; 26:5188-5197. [PMID: 32631957 DOI: 10.1158/1078-0432.ccr-20-0538] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/10/2020] [Accepted: 06/30/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Pretreatment selection of patients with non-small cell lung cancer (NSCLC) who would derive clinical benefit from treatment with immune checkpoint inhibitors (CPIs) would fulfill an unmet clinical need by reducing unnecessary toxicities from treatment and result in substantial health care savings. EXPERIMENTAL DESIGN In a retrospective study, mass spectrometry (MS)-based proteomic analysis was performed on pretreatment sera derived from patients with advanced NSCLC treated with nivolumab as part of routine clinical care (n = 289). Machine learning combined spectral and clinical data to stratify patients into three groups with good ("sensitive"), intermediate, and poor ("resistant") outcomes following treatment in the second-line setting. The test was applied to three independent patient cohorts and its biology was investigated using protein set enrichment analyses (PSEA). RESULTS A signature consisting of 274 MS features derived from a development set of 116 patients was associated with progression-free survival (PFS) and overall survival (OS) across two validation cohorts (N = 98 and N = 75). In pooled analysis, significantly better OS was demonstrated for "sensitive" relative to "not sensitive" patients treated with nivolumab; HR, 0.58 (95% confidence interval, 0.38-0-87; P = 0.009). There was no significant association with clinical factors including PD-L1 expression, available from 133 of 289 patients. The test demonstrated no significant association with PFS or OS in a historical cohort (n = 68) of second-line NSCLC patients treated with docetaxel. PSEA revealed proteomic classification to be significantly associated with complement and wound-healing cascades. CONCLUSIONS This serum-derived protein signature successfully stratified outcomes in cohorts of patients with advanced NSCLC treated with second-line PD-1 CPIs and deserves further prospective study.
Collapse
Affiliation(s)
- Mirte Muller
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Karlijn Hummelink
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daan P Hurkmans
- Department of Pulmonary Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Anna-Larissa N Niemeijer
- Department of Pulmonary Diseases, Vrije Universiteit VU Medical Center, Amsterdam, the Netherlands
| | - Kim Monkhorst
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | | | - Joachim G Aerts
- Department of Pulmonary Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Egbert F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Mass Spectrometry-Based Multivariate Proteomic Tests for Prediction of Outcomes on Immune Checkpoint Blockade Therapy: The Modern Analytical Approach. Int J Mol Sci 2020; 21:ijms21030838. [PMID: 32012941 PMCID: PMC7036840 DOI: 10.3390/ijms21030838] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
The remarkable success of immune checkpoint inhibitors (ICIs) has given hope of cure for some patients with advanced cancer; however, the fraction of responding patients is 15-35%, depending on tumor type, and the proportion of durable responses is even smaller. Identification of biomarkers with strong predictive potential remains a priority. Until now most of the efforts were focused on biomarkers associated with the assumed mechanism of action of ICIs, such as levels of expression of programmed death-ligand 1 (PD-L1) and mutation load in tumor tissue, as a proxy of immunogenicity; however, their performance is unsatisfactory. Several assays designed to capture the complexity of the disease by measuring the immune response in tumor microenvironment show promise but still need validation in independent studies. The circulating proteome contains an additional layer of information characterizing tumor-host interactions that can be integrated into multivariate tests using modern machine learning techniques. Here we describe several validated serum-based proteomic tests and their utility in the context of ICIs. We discuss test performances, demonstrate their independence from currently used biomarkers, and discuss various aspects of associated biological mechanisms. We propose that serum-based multivariate proteomic tests add a missing piece to the puzzle of predicting benefit from ICIs.
Collapse
|