1
|
Song P, Zhou D, Wang F, Li G, Bai L, Su J. Programmable biomaterials for bone regeneration. Mater Today Bio 2024; 29:101296. [PMID: 39469314 PMCID: PMC11513843 DOI: 10.1016/j.mtbio.2024.101296] [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: 07/12/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Programmable biomaterials are distinguished by their ability to adjust properties and functions on demand, in a periodic, reversible, or sequential manner. This contrasts with traditional biomaterials, which undergo irreversible, uncontrolled changes. This review synthesizes key advances in programmable biomaterials, examining their design principles, functionalities and applications in bone regeneration. It charts the transition from traditional to programmable biomaterials, emphasizing their enhanced precision, safety and control, which are critical from clinical and biosafety standpoints. We then classify programmable biomaterials into six types: dynamic nucleic acid-based biomaterials, electrically responsive biomaterials, bioactive scaffolds with programmable properties, nanomaterials for targeted bone regeneration, surface-engineered implants for sequential regeneration and stimuli-responsive release materials. Each category is analyzed for its structural properties and its impact on bone tissue engineering. Finally, the review further concludes by highlighting the challenges faced by programmable biomaterials and suggests integrating artificial intelligence and precision medicine to enhance their application in bone regeneration and other biomedical fields.
Collapse
Affiliation(s)
- Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Dongyang Zhou
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fuxiao Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghaizhongye Hospital, Shanghai, 200941, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
2
|
Nguyen JT, Jessri M, Costa-da-Silva AC, Sharma R, Mays JW, Treister NS. Oral Chronic Graft-Versus-Host Disease: Pathogenesis, Diagnosis, Current Treatment, and Emerging Therapies. Int J Mol Sci 2024; 25:10411. [PMID: 39408739 PMCID: PMC11476840 DOI: 10.3390/ijms251910411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Chronic graft-versus-host disease (cGvHD) is a multisystem disorder that occurs in recipients of allogeneic hematopoietic (alloHCT) stem cell transplants and is characterized by both inflammatory and fibrotic manifestations. It begins with the recognition of host tissues by the non-self (allogeneic) graft and progresses to tissue inflammation, organ dysfunction and fibrosis throughout the body. Oral cavity manifestations of cGVHD include mucosal features, salivary gland dysfunction and fibrosis. This review synthesizes current knowledge on the pathogenesis, diagnosis and management of oral cGVHD, with a focus on emerging trends and novel therapeutics. Data from various clinical studies and expert consensus are integrated to provide a comprehensive overview.
Collapse
Affiliation(s)
- Joe T. Nguyen
- Nguyen Laboratory, Head and Neck Cancer Section, Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Maryam Jessri
- Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD 4029, Australia;
- Department of Oral Medicine and Pathology, School of Dentistry, The University of Queensland, Herston, QLD 4072, Australia
| | - Ana C. Costa-da-Silva
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Rubina Sharma
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Jacqueline W. Mays
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA; (A.C.C.-d.-S.); (R.S.); (J.W.M.)
| | - Nathaniel S. Treister
- Division of Oral Medicine and Dentistry, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02114, USA
| |
Collapse
|
3
|
Canderan G, Muehling LM, Kadl A, Ladd S, Bonham C, Cross CE, Lima SM, Yin X, Sturek JM, Wilson JM, Keshavarz B, Bryant N, Murphy DD, Cheon IS, McNamara CA, Sun J, Utz PJ, Dolatshahi S, Irish JM, Woodfolk JA. Distinct Type 1 Immune Networks Underlie the Severity of Restrictive Lung Disease after COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587929. [PMID: 38617217 PMCID: PMC11014603 DOI: 10.1101/2024.04.03.587929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The variable etiology of persistent breathlessness after COVID-19 have confounded efforts to decipher the immunopathology of lung sequelae. Here, we analyzed hundreds of cellular and molecular features in the context of discrete pulmonary phenotypes to define the systemic immune landscape of post-COVID lung disease. Cluster analysis of lung physiology measures highlighted two phenotypes of restrictive lung disease that differed by their impaired diffusion and severity of fibrosis. Machine learning revealed marked CCR5+CD95+ CD8+ T-cell perturbations in mild-to-moderate lung disease, but attenuated T-cell responses hallmarked by elevated CXCL13 in more severe disease. Distinct sets of cells, mediators, and autoantibodies distinguished each restrictive phenotype, and differed from those of patients without significant lung involvement. These differences were reflected in divergent T-cell-based type 1 networks according to severity of lung disease. Our findings, which provide an immunological basis for active lung injury versus advanced disease after COVID-19, might offer new targets for treatment.
Collapse
|
4
|
Zhou Y, Smith J, Keerthi D, Li C, Sun Y, Mothi SS, Shyr DC, Spitzer B, Harris A, Chatterjee A, Chatterjee S, Shouval R, Naik S, Bertaina A, Boelens JJ, Triplett BM, Tang L, Sharma A. Longitudinal clinical data improve survival prediction after hematopoietic cell transplantation using machine learning. Blood Adv 2024; 8:686-698. [PMID: 37991991 PMCID: PMC10844815 DOI: 10.1182/bloodadvances.2023011752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 11/24/2023] Open
Abstract
ABSTRACT Serial prognostic evaluation after allogeneic hematopoietic cell transplantation (allo-HCT) might help identify patients at high risk of lethal organ dysfunction. Current prediction algorithms based on models that do not incorporate changes to patients' clinical condition after allo-HCT have limited predictive ability. We developed and validated a robust risk-prediction algorithm to predict short- and long-term survival after allo-HCT in pediatric patients that includes baseline biological variables and changes in the patients' clinical status after allo-HCT. The model was developed using clinical data from children and young adults treated at a single academic quaternary-care referral center. The model was created using a randomly split training data set (70% of the cohort), internally validated (remaining 30% of the cohort) and then externally validated on patient data from another tertiary-care referral center. Repeated clinical measurements performed from 30 days before allo-HCT to 30 days afterwards were extracted from the electronic medical record and incorporated into the model to predict survival at 100 days, 1 year, and 2 years after allo-HCT. Naïve-Bayes machine learning models incorporating longitudinal data were significantly better than models constructed from baseline variables alone at predicting whether patients would be alive or deceased at the given time points. This proof-of-concept study demonstrates that unlike traditional prognostic tools that use fixed variables for risk assessment, incorporating dynamic variability using clinical and laboratory data improves the prediction of mortality in patients undergoing allo-HCT.
Collapse
Affiliation(s)
- Yiwang Zhou
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jesse Smith
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Dinesh Keerthi
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Cai Li
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yilun Sun
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Suraj Sarvode Mothi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - David C. Shyr
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | - Barbara Spitzer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew Harris
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Avijit Chatterjee
- Digital, Informatics and Technology Solutions, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Subrata Chatterjee
- Digital, Informatics and Technology Solutions, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | - Jaap Jan Boelens
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Brandon M. Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Li Tang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
5
|
Baumrin E, Loren AW, Falk SJ, Mays JW, Cowen EW. Chronic graft-versus-host disease. Part I: Epidemiology, pathogenesis, and clinical manifestations. J Am Acad Dermatol 2024; 90:1-16. [PMID: 36572065 PMCID: PMC10287844 DOI: 10.1016/j.jaad.2022.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Chronic graft-versus-host disease is a major complication of allogeneic hematopoietic cell transplantation and a leading cause of long-term morbidity, nonrelapse mortality, and impaired health-related quality of life. The skin is commonly affected and presents heterogeneously, making the role of dermatologists critical in both diagnosis and treatment. In addition, new clinical classification and grading schemes inform treatment algorithms, which now include 3 U.S. Food and Drug Administration-approved therapies, and evolving transplant techniques are changing disease epidemiology. Part I reviews the epidemiology, pathogenesis, clinical manifestations, and diagnosis of chronic graft-versus-host disease. Part II discusses disease grading and therapeutic management.
Collapse
Affiliation(s)
- Emily Baumrin
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Alison W Loren
- Blood and Marrow Transplant, Cell Therapy and Transplant Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sandy J Falk
- Adult Survivorship Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Jacqueline W Mays
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Edward W Cowen
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
6
|
Niazi SK. Anti-Idiotypic mRNA Vaccine to Treat Autoimmune Disorders. Vaccines (Basel) 2023; 12:9. [PMID: 38276668 PMCID: PMC10819008 DOI: 10.3390/vaccines12010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/01/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
The 80+ existing autoimmune disorders (ADs) affect billions with little prevention or treatment options, except for temporary symptomatic management, leading to enormous human suffering and a monumental financial burden. The autoantibodies formed in most ADs have been identified, allowing the development of novel anti-idiotypic antibodies to mute the autoantibodies using vaccines. Nucleoside vaccines have been successfully tested as antigen-specific immunotherapies (ASI), with mRNA technology offering multi-epitope targeting to mute multiple autoantibodies. This paper proposes using mRNA technology to produce anti-idiotypic antibodies with broad effectiveness in preventing and treating them. This paper delves into the state-of-the-art mRNA design strategies used to develop novel ASIs by selecting appropriate T cell and B cell epitopes to generate anti-idiotypic antibodies. The low cost and fast development of mRNA vaccines make this technology the most affordable for the global control of ADs.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60012, USA
| |
Collapse
|
7
|
Buxbaum NP, Socié G, Hill GR, MacDonald KPA, Tkachev V, Teshima T, Lee SJ, Ritz J, Sarantopoulos S, Luznik L, Zeng D, Paczesny S, Martin PJ, Pavletic SZ, Schultz KR, Blazar BR. Chronic GvHD NIH Consensus Project Biology Task Force: evolving path to personalized treatment of chronic GvHD. Blood Adv 2023; 7:4886-4902. [PMID: 36322878 PMCID: PMC10463203 DOI: 10.1182/bloodadvances.2022007611] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 01/26/2023] Open
Abstract
Chronic graft-versus-host disease (cGvHD) remains a prominent barrier to allogeneic hematopoietic stem cell transplantion as the leading cause of nonrelapse mortality and significant morbidity. Tremendous progress has been achieved in both the understanding of pathophysiology and the development of new therapies for cGvHD. Although our field has historically approached treatment from an empiric position, research performed at the bedside and bench has elucidated some of the complex pathophysiology of cGvHD. From the clinical perspective, there is significant variability of disease manifestations between individual patients, pointing to diverse biological underpinnings. Capitalizing on progress made to date, the field is now focused on establishing personalized approaches to treatment. The intent of this article is to concisely review recent knowledge gained and formulate a path toward patient-specific cGvHD therapy.
Collapse
Affiliation(s)
- Nataliya P. Buxbaum
- Department of Pediatrics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Gerard Socié
- Hematology-Transplantation, Assistance Publique-Hopitaux de Paris & University of Paris – INSERM UMR 676, Hospital Saint Louis, Paris, France
| | - Geoffrey R. Hill
- Division of Medical Oncology, The University of Washington, Seattle, WA
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Kelli P. A. MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Victor Tkachev
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Stephanie J. Lee
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jerome Ritz
- Dana-Farber Cancer Institute, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA
| | - Stefanie Sarantopoulos
- Department of Medicine, Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Duke Cancer Institute, Durham, NC
| | - Leo Luznik
- Division of Hematologic Malignancies, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Defu Zeng
- Arthur D. Riggs Diabetes and Metabolism Research Institute, The Beckman Research Institute, Hematologic Maligancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, CA
| | - Sophie Paczesny
- Department of Microbiology and Immunology and Cancer Immunology Program, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Paul J. Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Steven Z. Pavletic
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kirk R. Schultz
- Michael Cuccione Childhood Cancer Research Program, British Columbia Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneappolis, MN
| |
Collapse
|
8
|
Baumrin E, Baker LX, Byrne M, Martin PJ, Flowers ME, Onstad L, El Jurdi N, Chen H, Beeghly-Fadiel A, Lee SJ, Tkaczyk ER. Prognostic Value of Cutaneous Disease Severity Estimates on Survival Outcomes in Patients With Chronic Graft-vs-Host Disease. JAMA Dermatol 2023; 159:393-402. [PMID: 36884224 PMCID: PMC9996455 DOI: 10.1001/jamadermatol.2022.6624] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/28/2022] [Indexed: 03/09/2023]
Abstract
Importance Prior studies have demonstrated an association between cutaneous chronic graft-vs-host disease (cGVHD) and mortality. Assessment of the prognostic value of different measures of disease severity would assist in risk stratification. Objective To compare the prognostic value of body surface area (BSA) and National Institutes of Health (NIH) Skin Score on survival outcomes stratified by erythema and sclerosis subtypes of cGVHD. Design, Setting, and Participants Multicenter prospective cohort study from the Chronic Graft-vs-Host Disease Consortium including 9 medical centers in the US, enrolled from 2007 through 2012 and followed until 2018. Participants were adults and children with a diagnosis of cGVHD requiring systemic immunosuppression and with skin involvement during the study period, who had longitudinal follow-up. Data analysis was performed from April 2019 to April 2022. Exposures Patients underwent continuous BSA estimation and categorical NIH Skin Score grading of cutaneous cGVHD at enrollment and every 3 to 6 months thereafter. Main Outcomes and Measures Nonrelapse mortality (NRM) and overall survival (OS), compared between BSA and NIH Skin Score longitudinal prognostic models, adjusted for age, race, conditioning intensity, patient sex, and donor sex. Results Of 469 patients with cGVHD, 267 (57%) (105 female [39%]; mean [SD] age, 51 [12] years) had cutaneous cGVHD at enrollment, and 89 (19%) developed skin involvement subsequently. Erythema-type disease had earlier onset and was more responsive to treatment compared with sclerosis-type disease. Most cases (77 of 112 [69%]) of sclerotic disease occurred without prior erythema. Erythema-type cGVHD at first follow-up visit was associated with NRM (hazard ratio, 1.33 per 10% BSA increase; 95% CI, 1.19-1.48; P < .001) and OS (hazard ratio, 1.28 per 10% BSA increase; 95% CI, 1.14-1.44; P < .001), while sclerosis-type cGVHD had no significant association with mortality. The model with erythema BSA collected at baseline and first follow-up visits retained 75% of the total prognostic information (from all covariates including BSA and NIH Skin Score) for NRM and 73% for OS, with no statistical difference between prognostic models (likelihood ratio test χ2, 5.9; P = .05). Conversely, NIH Skin Score collected at the same intervals lost significant prognostic information (likelihood ratio test χ2, 14.7; P < .001). The model incorporating NIH Skin Score instead of erythema BSA accounted for only 38% of the total information for NRM and 58% for OS. Conclusions and Relevance In this prospective cohort study, erythema-type cutaneous cGVHD was associated with increased risk of mortality. Erythema BSA collected at baseline and follow-up predicted survival more accurately than the NIH Skin Score in patients requiring immunosuppression. Accurate assessment of erythema BSA may assist in identifying patients with cutaneous cGVHD at high risk for mortality.
Collapse
Affiliation(s)
- Emily Baumrin
- Department of Dermatology, University of Pennsylvania, Philadelphia
| | - Laura X. Baker
- Department of Dermatology, University of California, San Francisco
| | - Michael Byrne
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paul J. Martin
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle
| | - Mary E. Flowers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle
| | - Lynn Onstad
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Najla El Jurdi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Stephanie J. Lee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle
| | - Eric R. Tkaczyk
- Department of Veterans Affairs, Nashville, Tennessee
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
9
|
Booth G, Yu Y, Harlan RP, Jacoby CE, Tomic KM, Slater SE, Allen BE, Berklich EM, Knight RJ, Dela Cruz J, Fu R, Gandhi A, Cook RJ, Meyers G, Maziarz RT, Newell LF. Day 4 collection of granulocyte colony-stimulating factor-mobilized HLA-matched sibling donor peripheral blood allografts demonstrates no long-term increase in chronic graft-versus-host disease or relapse rates. Cytotherapy 2023; 25:423-431. [PMID: 36690537 DOI: 10.1016/j.jcyt.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND AIMS In a previous pilot study of HLA-matched sibling donor hematopoietic cell transplantation (HCT), the authors determined the feasibility of day 4 versus day 5 granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood stem cell (PBSC) collection compared with a historical cohort. Given identified differences in the PBSC product (day 4 cohort with significantly lower infused total nucleated, mononuclear and CD3 cells compared with other collection cohorts), the authors performed a follow-up study to determine long-term post-HCT outcomes, including detailed characterization of chronic graft-versus-host disease (GVHD). METHODS This was a prospective observational study, and the authors collected data on chronic GVHD, staging, sites of involvement and treatments. Performance status, incidence of relapse, overall survival and duration of immunosuppressive therapy (IST) were also evaluated. Data were examined retrospectively. To account for differences in length of follow-up among cohorts, the authors also determined performance status and chronic GVHD staging, sites and treatment at 2 years post-HCT. RESULTS At 2 years post-HCT, the overall survival rate was 71.7% in the day 4 cohort compared with 61.5%, 52% and 56% in the day 5, 2-day and historical cohorts, respectively (P = 0.283). The cumulative incidence of chronic GVHD was 65.2% in the day 4 cohort versus 46.4% in the day 5 cohort, 51.1% in the 2-day cohort and 65% in the historical cohort (P = 0.26). There was no significant difference in the maximum overall stage of chronic GVHD (P = 0.513), median number of sites involved (P = 0.401) or cumulative incidence of discontinuation of IST (P = 0.32). Death from chronic GVHD was less common in the day 4 and day 5 cohorts compared with the 2-day and historical cohorts, though this did not reach statistical significance. CONCLUSIONS The authors' preliminary results demonstrated that collection of allogeneic matched sibling donor PBSCs on day 4 of G-CSF was feasible, reduced donor exposure to growth factor and was associated with an initial cost savings. Importantly, the authors now demonstrate that transplantation of day 4 mobilized PBSCs is not associated with any adverse outcomes post-HCT, including late effects such as chronic GVHD. Further investigation of donor G-CSF collection algorithms is merited in other HCT settings, including unrelated and mismatched related donors.
Collapse
Affiliation(s)
- Georgeann Booth
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Yun Yu
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rogelyn P Harlan
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Carol E Jacoby
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Kaitlyn M Tomic
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Susan E Slater
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Bryon E Allen
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Elizabeth M Berklich
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rebekah J Knight
- Cellular Therapy Laboratory, Hospital and Clinics, Oregon Health & Science University, Portland
| | - Julieann Dela Cruz
- Cellular Therapy Laboratory, Hospital and Clinics, Oregon Health & Science University, Portland
| | - Rongwei Fu
- Biostatistics Shared Resources, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA; Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University-Portland State University School of Public Health, Oregon Health and Science University, Portland, Oregon, USA
| | - Arpita Gandhi
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rachel J Cook
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Gabrielle Meyers
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Richard T Maziarz
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Laura F Newell
- Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|
10
|
Zelić Kerep A, Olivieri A, Schoemans H, Lawitschka A, Halter J, Pulanic D, Dickinson A, Greinix HT, Pavletic SZ, Schultz KR, Lee SJ, Wolff D. Chronic gvhd dictionary-eurograft cost action initiative consensus report. Bone Marrow Transplant 2023; 58:68-71. [PMID: 36229646 DOI: 10.1038/s41409-022-01837-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/05/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023]
Abstract
Chronic graft versus host disease (cGVHD) affects patients after allogeneic hematopoietic stem cell transplantation (alloHSCT). This orphan disease poses a challenge for clinicians and researchers. The purpose of the cGVHD Dictionary is to provide a standardized structure for cGVHD databases on an international level, reconciling differences in data retrieval and facilitate database merging. It is derived from several consensus meetings of the EUROGRAFT consortium (European Cooperation in Science and Technology-COST Action CA17138) followed by a consensus process involving European Society for Blood and Marrow Transplantation (EBMT), US GvHD consortium and Center for International Bone Marrow Transplant Registry (CIBMTR). Databases used for the dictionary were: the National Institutes of Health (NIH) database, the Center for International Blood and Marrow Transplant Research, Applying Biomarkers to Minimize Long Term Effects of Childhood/Adolescent Cancer Treatment - Pediatric Blood and Marrow Transplant Consortium database, EBMT registry, the German-Austrian-Swiss GvHD registry, Italian Blood and Marrow Transplantation Society registry and Regensburg-Göttingen-Newcastle HSCT dataset. A four-part cGVHD Dictionary was formed based on the databases, consensus, and evidence in the literature. The Dictionary is divided into: (1) Patient characteristics, (2) Transplant characteristics, (3) cGVHD characteristics and (4) patient-reported quality of life, symptom burden and functional indicators.
Collapse
Affiliation(s)
- Ana Zelić Kerep
- Division of Hematology, Department of Internal Medicine, University Hospital Center Zagreb, Zagreb, Croatia.
| | - Atillio Olivieri
- Department of Clinical and Molecular Sciences, University of Ancona, Ancona, Italy
| | - Helene Schoemans
- Department of Hematology, University Hospital Leuven, Leuven, Belgium.,Deparment of Public Health and Primary Care, ACCENT VV, KU Leuven-University of Leuven, Leuven, Belgium
| | - Anita Lawitschka
- Stem Cell Transplant Unit, St. Anna Children's Hospital, Medical University Vienna, Vienna, Austria
| | - Jörg Halter
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Drazen Pulanic
- Division of Hematology, Department of Internal Medicine, University Hospital Center Zagreb, Zagreb, Croatia.,University of Zagreb School of Medicine, Zagreb, Croatia
| | - Anne Dickinson
- Hematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle, UK
| | | | - Steven Z Pavletic
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kirk R Schultz
- Division of Hematology/Oncology/BMT, British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada
| | | | - Daniel Wolff
- Department of Internal Medicine III, University of Regensburg, Regensburg, Germany
| |
Collapse
|
11
|
Wongvibulsin S, Frech TM, Chren MM, Tkaczyk ER. Expanding Personalized, Data-Driven Dermatology: Leveraging Digital Health Technology and Machine Learning to Improve Patient Outcomes. JID INNOVATIONS 2022; 2:100105. [PMID: 35462957 PMCID: PMC9026581 DOI: 10.1016/j.xjidi.2022.100105] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/13/2021] [Accepted: 01/07/2022] [Indexed: 11/30/2022] Open
Abstract
The current revolution of digital health technology and machine learning offers enormous potential to improve patient care. Nevertheless, it is essential to recognize that dermatology requires an approach different from those of other specialties. For many dermatological conditions, there is a lack of standardized methodology for quantitatively tracking disease progression and treatment response (clinimetrics). Furthermore, dermatological diseases impact patients in complex ways, some of which can be measured only through patient reports (psychometrics). New tools using digital health technology (e.g., smartphone applications, wearable devices) can aid in capturing both clinimetric and psychometric variables over time. With these data, machine learning can inform efforts to improve health care by, for example, the identification of high-risk patient groups, optimization of treatment strategies, and prediction of disease outcomes. We use the term personalized, data-driven dermatology to refer to the use of comprehensive data to inform individual patient care and improve patient outcomes. In this paper, we provide a framework that includes data from multiple sources, leverages digital health technology, and uses machine learning. Although this framework is applicable broadly to dermatological conditions, we use the example of a serious inflammatory skin condition, chronic cutaneous graft-versus-host disease, to illustrate personalized, data-driven dermatology.
Collapse
Affiliation(s)
- Shannon Wongvibulsin
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Tracy M. Frech
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, Tennessee, USA
| | - Mary-Margaret Chren
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Eric R. Tkaczyk
- VA Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, Tennessee, USA
- Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
12
|
Lee S, Lee E, Park SS, Park MS, Jung J, Min GJ, Park S, Lee SE, Cho BS, Eom KS, Kim YJ, Lee S, Kim HJ, Min CK, Cho SG, Lee JW, Hwang HJ, Yoon JH. Prediction and recommendation by machine learning through repetitive internal validation for hepatic veno-occlusive disease/sinusoidal obstruction syndrome and early death after allogeneic hematopoietic cell transplantation. Bone Marrow Transplant 2022; 57:538-546. [PMID: 35075247 DOI: 10.1038/s41409-022-01583-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/23/2022]
Abstract
Using traditional statistical methods, we previously analyzed the risk factors and treatment outcomes of veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS) after allogeneic hematopoietic cell transplantation. Within the same cohort, we applied machine learning to create prediction and recommendation models. We analyzed 2572 transplants using eXtreme Gradient Boosting (XGBoost) to predict post-transplant VOD/SOS and early death. Using the XGBoost and SHapley Additive exPlanations (SHAP), we found influential factors and devised recommendation models, which were internally verified by repetitive ten-fold cross-validation. SHAP values suggested that gender, busulfan dosage, age, forced expiratory volume, and Disease Risk Index were significant factors for VOD/SOS. The areas under the receiver operating characteristic curves and the areas under the precision-recall curve of the models were 0.740, 0.144 for all VOD/SOS, 0.793, 0.793 for severe to very severe VOD/SOS, and 0.746, 0.304 for early death. According to our single feature recommendation, following the busulfan dosage was the most effective for preventing VOD/SOS. The recommendation method for six adjustable feature sets was also validated, and a subgroup corresponding to five to six features showed significant preventive power for VOD/SOS and early death. Our personalized treatment set recommendation showed reproducibility in repetitive internal validation, but large external cohorts should prospectively validate our model.
Collapse
Affiliation(s)
| | - Eunsaem Lee
- Department of Mathematics, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Korea
| | - Sung-Soo Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Sue Park
- Department of Mathematics, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Korea
| | | | - Gi June Min
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Silvia Park
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Eun Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung-Sik Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki-Seong Eom
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoo-Jin Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee-Je Kim
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang-Ki Min
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok-Goo Cho
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Wook Lee
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyung Ju Hwang
- AMSquare Corp., Pohang, Gyeongbuk, Korea.
- Department of Mathematics, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Korea.
| | - Jae-Ho Yoon
- Department of Hematology, Catholic Hematology Hospital and Leukemia Research Institute, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
13
|
Artificial Intelligence in Clinical Immunology. Artif Intell Med 2022. [DOI: 10.1007/978-3-030-64573-1_83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
14
|
Performance evaluation of machine learning for breast cancer diagnosis: A case study. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.101009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
15
|
Zaccaria GM, Ferrero S, Hoster E, Passera R, Evangelista A, Genuardi E, Drandi D, Ghislieri M, Barbero D, Del Giudice I, Tani M, Moia R, Volpetti S, Cabras MG, Di Renzo N, Merli F, Vallisa D, Spina M, Pascarella A, Latte G, Patti C, Fabbri A, Guarini A, Vitolo U, Hermine O, Kluin-Nelemans HC, Cortelazzo S, Dreyling M, Ladetto M. A Clinical Prognostic Model Based on Machine Learning from the Fondazione Italiana Linfomi (FIL) MCL0208 Phase III Trial. Cancers (Basel) 2021; 14:188. [PMID: 35008361 PMCID: PMC8750124 DOI: 10.3390/cancers14010188] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/26/2021] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Multicenter clinical trials are producing growing amounts of clinical data. Machine Learning (ML) might facilitate the discovery of novel tools for prognostication and disease-stratification. Taking advantage of a systematic collection of multiple variables, we developed a model derived from data collected on 300 patients with mantle cell lymphoma (MCL) from the Fondazione Italiana Linfomi-MCL0208 phase III trial (NCT02354313). METHODS We developed a score with a clustering algorithm applied to clinical variables. The candidate score was correlated to overall survival (OS) and validated in two independent data series from the European MCL Network (NCT00209222, NCT00209209); Results: Three groups of patients were significantly discriminated: Low, Intermediate (Int), and High risk (High). Seven discriminants were identified by a feature reduction approach: albumin, Ki-67, lactate dehydrogenase, lymphocytes, platelets, bone marrow infiltration, and B-symptoms. Accordingly, patients in the Int and High groups had shorter OS rates than those in the Low and Int groups, respectively (Int→Low, HR: 3.1, 95% CI: 1.0-9.6; High→Int, HR: 2.3, 95% CI: 1.5-4.7). Based on the 7 markers, we defined the engineered MCL international prognostic index (eMIPI), which was validated and confirmed in two independent cohorts; Conclusions: We developed and validated a ML-based prognostic model for MCL. Even when currently limited to baseline predictors, our approach has high scalability potential.
Collapse
Affiliation(s)
- Gian Maria Zaccaria
- Unit of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (S.F.); (E.G.); (D.D.); (D.B.)
- Unit of Hematology and Cell Therapy, IRCCS-Istituto Tumori ‘Giovanni Paolo II’, 70124 Bari, Italy;
| | - Simone Ferrero
- Unit of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (S.F.); (E.G.); (D.D.); (D.B.)
| | - Eva Hoster
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany;
| | - Roberto Passera
- Division of Nuclear Medicine, University of Torino, 10126 Turin, Italy;
| | - Andrea Evangelista
- Unit of Clinical Epidemiology, CPO Piemonte, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy;
| | - Elisa Genuardi
- Unit of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (S.F.); (E.G.); (D.D.); (D.B.)
| | - Daniela Drandi
- Unit of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (S.F.); (E.G.); (D.D.); (D.B.)
| | - Marco Ghislieri
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129 Turin, Italy;
- PoliToBIOMedLab of Politecnico di Torino, 10129 Turin, Italy
| | - Daniela Barbero
- Unit of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (S.F.); (E.G.); (D.D.); (D.B.)
| | - Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Monica Tani
- Hematology Unit, Santa Maria delle Croci Hospital, 48121 Ravenna, Italy;
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy; (R.M.); (M.L.)
| | - Stefano Volpetti
- Unit of Hematology, Presidio Ospedaliero Universitario “Santa Maria della Misericordia”, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy;
| | | | - Nicola Di Renzo
- Unit of Hematology and Bone Marrow Transplant, ‘V. Fazzi’ Hospital, 73100 Lecce, Italy;
| | | | - Daniele Vallisa
- Unit of Hematology, Department of Oncology and Hematology, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
| | - Michele Spina
- Division of Medical Oncology and Immune-Related Tumors, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Anna Pascarella
- Unit of Hematology, dell’ Angelo Mestre-Venezia Hospital, 30174 Mestre-Venezia, Italy;
| | - Giancarlo Latte
- Unit of Hematology and Bone Marrow Transplant, ‘San Francesco’ Hospital, 08100 Nuoro, Italy;
| | - Caterina Patti
- Unit of Hematology, Azienda Ospedali Riuniti Villa Sofia-Cervello, 90146 Palermo, Italy;
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy;
| | - Attilio Guarini
- Unit of Hematology and Cell Therapy, IRCCS-Istituto Tumori ‘Giovanni Paolo II’, 70124 Bari, Italy;
| | - Umberto Vitolo
- Division of Hematology, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, 10126 Turin, Italy;
| | - Olivier Hermine
- Service D’hématologie, Hôpital Universitaire Necker, Université René Descartes, Assistance Publique Hôpitaux de Paris, 75015 Paris, France;
| | - Hanneke C Kluin-Nelemans
- Department of Haematology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands;
| | | | - Martin Dreyling
- Department of Medicine III, University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Marco Ladetto
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy; (R.M.); (M.L.)
- Division of Hematology, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| |
Collapse
|
16
|
Establishment of a Predictive Model for GvHD-free, Relapse-free Survival after Allogeneic HSCT using Ensemble Learning. Blood Adv 2021; 6:2618-2627. [PMID: 34933327 PMCID: PMC9043925 DOI: 10.1182/bloodadvances.2021005800] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/23/2021] [Indexed: 12/03/2022] Open
Abstract
Stacked ensemble of machine-learning algorithms could establish more accurate prediction model for survival analysis than existing methods. Stacked ensemble model can be applied to personalized prediction of HSCT outcomes from pretransplant characteristics.
Graft-versus-host disease-free, relapse-free survival (GRFS) is a useful composite end point that measures survival without relapse or significant morbidity after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We aimed to develop a novel analytical method that appropriately handles right-censored data and competing risks to understand the risk for GRFS and each component of GRFS. This study was a retrospective data-mining study on a cohort of 2207 adult patients who underwent their first allo-HSCT within the Kyoto Stem Cell Transplantation Group, a multi-institutional joint research group of 17 transplantation centers in Japan. The primary end point was GRFS. A stacked ensemble of Cox Proportional Hazard (Cox-PH) regression and 7 machine-learning algorithms was applied to develop a prediction model. The median age for the patients was 48 years. For GRFS, the stacked ensemble model achieved better predictive accuracy evaluated by C-index than other state-of-the-art competing risk models (ensemble model: 0.670; Cox-PH: 0.668; Random Survival Forest: 0.660; Dynamic DeepHit: 0.646). The probability of GRFS after 2 years was 30.54% for the high-risk group and 40.69% for the low-risk group (hazard ratio compared with the low-risk group: 2.127; 95% CI, 1.19-3.80). We developed a novel predictive model for survival analysis that showed superior risk stratification to existing methods using a stacked ensemble of multiple machine-learning algorithms.
Collapse
|
17
|
Nagy M, Radakovich N, Nazha A. Machine Learning in Oncology: What Should Clinicians Know? JCO Clin Cancer Inform 2021; 4:799-810. [PMID: 32926637 DOI: 10.1200/cci.20.00049] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The volume and complexity of scientific and clinical data in oncology have grown markedly over recent years, including but not limited to the realms of electronic health data, radiographic and histologic data, and genomics. This growth holds promise for a deeper understanding of malignancy and, accordingly, more personalized and effective oncologic care. Such goals require, however, the development of new methods to fully make use of the wealth of available data. Improvements in computer processing power and algorithm development have positioned machine learning, a branch of artificial intelligence, to play a prominent role in oncology research and practice. This review provides an overview of the basics of machine learning and highlights current progress and challenges in applying this technology to cancer diagnosis, prognosis, and treatment recommendations, including a discussion of current takeaways for clinicians.
Collapse
Affiliation(s)
- Matthew Nagy
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Nathan Radakovich
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Aziz Nazha
- Center for Clinical Artificial Intelligence, Cleveland Clinic, Cleveland, OH.,Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
18
|
Jung SM, Park KS, Kim KJ. Deep phenotyping of synovial molecular signatures by integrative systems analysis in rheumatoid arthritis. Rheumatology (Oxford) 2021; 60:3420-3431. [PMID: 33230538 DOI: 10.1093/rheumatology/keaa751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/29/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE RA encompasses a complex, heterogeneous and dynamic group of diseases arising from molecular and cellular perturbations of synovial tissues. The aim of this study was to decipher this complexity using an integrative systems approach and provide novel insights for designing stratified treatments. METHODS An RNA sequencing dataset of synovial tissues from 152 RA patients and 28 normal controls was imported and subjected to filtration of differentially expressed genes, functional enrichment and network analysis, non-negative matrix factorization, and key driver analysis. A naïve Bayes classifier was applied to the independent datasets to investigate the factors associated with treatment outcome. RESULTS A matrix of 1241 upregulated differentially expressed genes from RA samples was classified into three subtypes (C1-C3) with distinct molecular and cellular signatures. C3 with prominent immune cells and proinflammatory signatures had a stronger association with the presence of ACPA and showed a better therapeutic response than C1 and C2, which were enriched with neutrophil and fibroblast signatures, respectively. C2 was more occupied by synovial fibroblasts of destructive phenotype and carried highly expressed key effector molecules of invasion and osteoclastogenesis. CXCR2, JAK3, FYN and LYN were identified as key driver genes in C1 and C3. HDAC, JUN, NFKB1, TNF and TP53 were key regulators modulating fibroblast aggressiveness in C2. CONCLUSIONS Deep phenotyping of synovial heterogeneity captured comprehensive and discrete pathophysiological attributes of RA regarding clinical features and treatment response. This result could serve as a template for future studies to design stratified approaches for RA patients.
Collapse
Affiliation(s)
- Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| | - Kyung-Su Park
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ki-Jo Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
19
|
Barone SM, Paul AGA, Muehling LM, Lannigan JA, Kwok WW, Turner RB, Woodfolk JA, Irish JM. Unsupervised machine learning reveals key immune cell subsets in COVID-19, rhinovirus infection, and cancer therapy. eLife 2021; 10:e64653. [PMID: 34350827 PMCID: PMC8370768 DOI: 10.7554/elife.64653] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 08/02/2021] [Indexed: 12/31/2022] Open
Abstract
For an emerging disease like COVID-19, systems immunology tools may quickly identify and quantitatively characterize cells associated with disease progression or clinical response. With repeated sampling, immune monitoring creates a real-time portrait of the cells reacting to a novel virus before disease-specific knowledge and tools are established. However, single cell analysis tools can struggle to reveal rare cells that are under 0.1% of the population. Here, the machine learning workflow Tracking Responders EXpanding (T-REX) was created to identify changes in both rare and common cells across human immune monitoring settings. T-REX identified cells with highly similar phenotypes that localized to hotspots of significant change during rhinovirus and SARS-CoV-2 infections. Specialized MHCII tetramer reagents that mark rhinovirus-specific CD4+ cells were left out during analysis and then used to test whether T-REX identified biologically significant cells. T-REX identified rhinovirus-specific CD4+ T cells based on phenotypically homogeneous cells expanding by ≥95% following infection. T-REX successfully identified hotspots of virus-specific T cells by comparing infection (day 7) to either pre-infection (day 0) or post-infection (day 28) samples. Plotting the direction and degree of change for each individual donor provided a useful summary view and revealed patterns of immune system behavior across immune monitoring settings. For example, the magnitude and direction of change in some COVID-19 patients was comparable to blast crisis acute myeloid leukemia patients undergoing a complete response to chemotherapy. Other COVID-19 patients instead displayed an immune trajectory like that seen in rhinovirus infection or checkpoint inhibitor therapy for melanoma. The T-REX algorithm thus rapidly identifies and characterizes mechanistically significant cells and places emerging diseases into a systems immunology context for comparison to well-studied immune changes.
Collapse
Affiliation(s)
- Sierra M Barone
- Department of Cell and Developmental Biology, Vanderbilt UniversityNashvilleUnited States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical CenterNashvilleUnited States
| | - Alberta GA Paul
- Allergy Division, Department of Medicine, University of Virginia School of MedicineCharlottesvilleUnited States
| | - Lyndsey M Muehling
- Allergy Division, Department of Medicine, University of Virginia School of MedicineCharlottesvilleUnited States
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of MedicineCharlottesvilleUnited States
| | - Joanne A Lannigan
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of MedicineCharlottesvilleUnited States
| | - William W Kwok
- Benaroya Research Institute at Virginia MasonSeattleUnited States
| | - Ronald B Turner
- Department of Pediatrics, University of Virginia School of MedicineCharlottesvilleUnited States
| | - Judith A Woodfolk
- Allergy Division, Department of Medicine, University of Virginia School of MedicineCharlottesvilleUnited States
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of MedicineCharlottesvilleUnited States
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt UniversityNashvilleUnited States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical CenterNashvilleUnited States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical CenterNashvilleUnited States
| |
Collapse
|
20
|
Mackay BS, Marshall K, Grant-Jacob JA, Kanczler J, Eason RW, Oreffo ROC, Mills B. The future of bone regeneration: integrating AI into tissue engineering. Biomed Phys Eng Express 2021; 7. [PMID: 34271556 DOI: 10.1088/2057-1976/ac154f] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/16/2021] [Indexed: 01/16/2023]
Abstract
Tissue engineering is a branch of regenerative medicine that harnesses biomaterial and stem cell research to utilise the body's natural healing responses to regenerate tissue and organs. There remain many unanswered questions in tissue engineering, with optimal biomaterial designs still to be developed and a lack of adequate stem cell knowledge limiting successful application. Advances in artificial intelligence (AI), and deep learning specifically, offer the potential to improve both scientific understanding and clinical outcomes in regenerative medicine. With enhanced perception of how to integrate artificial intelligence into current research and clinical practice, AI offers an invaluable tool to improve patient outcome.
Collapse
Affiliation(s)
- Benita S Mackay
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Karen Marshall
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom
| | - James A Grant-Jacob
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Janos Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom
| | - Robert W Eason
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Institute of Developmental Sciences, Faculty of Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom.,Institute of Developmental Sciences, Faculty of Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Ben Mills
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| |
Collapse
|
21
|
Xue E, Lorentino F, Pavesi F, Assanelli A, Peccatori J, Bernardi M, Corti C, Ciceri F, Lupo Stanghellini MT. Ruxolitinib for chronic steroid-refractory graft versus host disease: a single center experience. Leuk Res 2021; 109:106642. [PMID: 34157510 DOI: 10.1016/j.leukres.2021.106642] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/05/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Chronic Graft versus Host Disease (GvHD) is a serious complication of allogeneic hematopoietic stem cell transplant that severely impacts quality of life and long-term survival. About 50-to-60 % of patients treated with steroids require a further line of therapy due to lack of sustained response. Ruxolitinib, a JAK1/2 inhibitor, has recently been approved for the treatment of acute GvHD. METHODS We aimed to retrospectively evaluate ruxolitinib efficacy and safety in a cohort of patients diagnosed with moderate (25 %) or severe (75 %) steroid-refractory or steroid-dependent chronic GvHD. Response evaluation was performed at three and six months. RESULTS Thirty-six patients received ruxolitinib after a median of three previous lines (range, r 1-11) for a median of 8.6 months (r 1-51.6). Cutaneous GvHD was the most frequent presentation. We observed an overall response of 59 % (CR 9%, PR 50 %) at three months and 62 % (CR 15 %, PR 46 %) at six months. Two patients had hematologic disease recurrence and were censored at relapse; no other permanent discontinuation due to adverse events were documented. Cutaneous, oral, genital and ocular GvHD significantly improved after treatment. 2-year overall survival and 2-year transplant related mortality were 74 % and 19 % respectively. Ruxolitinib was associated with a significant reduction of steroid dose. CONCLUSION Ruxolitinib was confirmed to be a safe and effective option as salvage treatment also for advanced stages of chronic GvHD. Longer follow up is needed to evaluate durability of response. Prospective analyses on larger cohorts are ongoing.
Collapse
Affiliation(s)
- Elisabetta Xue
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy
| | - Francesca Lorentino
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy; PhD Program in Public Health, School of Medicine and Surgery, University of Milano Bicocca, Italy
| | - Francesca Pavesi
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy
| | - Andrea Assanelli
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy
| | - Jacopo Peccatori
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy
| | - Massimo Bernardi
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy
| | - Consuelo Corti
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy
| | - Fabio Ciceri
- Haematology and Bone Marrow Transplant Unit, IRCSS San Raffaele Scientific Institute, Italy
| | | |
Collapse
|
22
|
Shakshouk H, Tkaczyk ER, Cowen EW, El-Azhary RA, Hashmi SK, Kenderian SJ, Lehman JS. Methods to Assess Disease Activity and Severity in Cutaneous Chronic Graft-versus-Host Disease: A Critical Literature Review. Transplant Cell Ther 2021; 27:738-746. [PMID: 34107339 DOI: 10.1016/j.jtct.2021.05.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/26/2022]
Abstract
Chronic graft-versus-host disease (cGVHD), a potentially debilitating complication of hematopoietic cell transplantation, confers increased risk for mortality. Whereas treatment decisions rely on an accurate assessment of disease activity/severity, validated methods of assessing cutaneous cGVHD activity/severity appear to be limited. In this study, we aimed to identify and evaluate current data on the assessment of disease activity/severity in cutaneous cGVHD. Using modified PRISMA methods, we performed a critical literature review for relevant articles. Our literature search identified 1741 articles, of which 1635 were excluded as duplicates or failure to meet inclusion criteria. Of the included studies (n = 106), 39 (37%) addressed clinical and/or histopathologic parameters, 53 (50%) addressed serologic parameters, 8 (7.5%) addressed imaging parameters, and 6 (5.5%) addressed computer-based technologies. The only formally validated metric of disease activity/severity assessment in cutaneous cGVHD is the National Institutes of Health consensus scoring system, which is founded on clinical assessment alone. The lack of an objective marker for cGVHD necessitates further studies. An evaluation of the potential contributions of serologic, imaging, and/or computer-based technologies is warranted.
Collapse
Affiliation(s)
- Hadir Shakshouk
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota; Department of Dermatology, Andrology and Venerology, Alexandria University, Alexandria, Egypt
| | - Eric R Tkaczyk
- Dermatology and Research Services, Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN; Department of Dermatology, Vanderbilt University Medical Center, Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Edward W Cowen
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Shahrukh K Hashmi
- Division of Hematology, Mayo Clinic, Rochester, Minnesota; Department of Medicine, Sheikh Shakhbout Medical City, Abu Dhabi, UAE
| | | | - Julia S Lehman
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
23
|
Kitko CL, Pidala J, Schoemans HM, Lawitschka A, Flowers ME, Cowen EW, Tkaczyk E, Farhadfar N, Jain S, Steven P, Luo ZK, Ogawa Y, Stern M, Yanik GA, Cuvelier GDE, Cheng GS, Holtan SG, Schultz KR, Martin PJ, Lee SJ, Pavletic SZ, Wolff D, Paczesny S, Blazar BR, Sarantopoulos S, Socie G, Greinix H, Cutler C. National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease: IIa. The 2020 Clinical Implementation and Early Diagnosis Working Group Report. Transplant Cell Ther 2021; 27:545-557. [PMID: 33839317 PMCID: PMC8803210 DOI: 10.1016/j.jtct.2021.03.033] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022]
Abstract
Recognition of the earliest signs and symptoms of chronic graft-versus-host disease (GVHD) that lead to severe manifestations remains a challenge. The standardization provided by the National Institutes of Health (NIH) 2005 and 2014 consensus projects has helped improve diagnostic accuracy and severity scoring for clinical trials, but utilization of these tools in routine clinical practice is variable. Additionally, when patients meet the NIH diagnostic criteria, many already have significant morbidity and possibly irreversible organ damage. The goals of this early diagnosis project are 2-fold. First, we provide consensus recommendations regarding implementation of the current NIH diagnostic guidelines into routine transplant care, outside of clinical trials, aiming to enhance early clinical recognition of chronic GVHD. Second, we propose directions for future research efforts to enable discovery of new, early laboratory as well as clinical indicators of chronic GVHD, both globally and for highly morbid organ-specific manifestations. Identification of early features of chronic GVHD that have high positive predictive value for progression to more severe manifestations of the disease could potentially allow for future pre-emptive clinical trials.
Collapse
Affiliation(s)
- Carrie L Kitko
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Joseph Pidala
- Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Hélène M Schoemans
- Department of Hematology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Anita Lawitschka
- St. Anna Children's Hospital, Children's Cancer Research Institute, Vienna, Austria
| | - Mary E Flowers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Edward W Cowen
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland
| | - Eric Tkaczyk
- Research & Dermatology Services, Department of Veterans Affairs, Nashville, Tennessee; Vanderbilt Dermatology Translational Research Clinic, Department of Dermatology, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Nosha Farhadfar
- Division of Hematology/Oncology, University of Florida College of Medicine, Gainesville, Florida
| | - Sandeep Jain
- Department of Ophthalmology, University of Illinois at Chicago, Chicago, Illinois
| | - Philipp Steven
- Division for Dry-Eye Disease and Ocular GVHD, Department of Ophthalmology, Medical Faculty and University Hospital, University of Cologne, Cologne, Germany
| | - Zhonghui K Luo
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard University, Boston, Massachusetts
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Michael Stern
- Department of Ophthalmology, University of Illinois at Chicago, Chicago, Illinois; ImmunEyez LLC, Irvine, California
| | - Greg A Yanik
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Geoffrey D E Cuvelier
- Pediatric Blood and Marrow Transplantation, Department of Pediatric Oncology-Hematology-BMT, CancerCare Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Guang-Shing Cheng
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Shernan G Holtan
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Kirk R Schultz
- Pediatric Hematology/Oncology/BMT, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Paul J Martin
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Steven Z Pavletic
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Daniel Wolff
- Department of Internal Medicine III, University Hospital of Regensburg, Regensburg, Germany
| | - Sophie Paczesny
- Department of Microbiology and Immunology, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, Minnesota
| | - Stephanie Sarantopoulos
- Division of Hematological Malignancies and Cellular Therapy, Duke University Department of Medicine, Duke Cancer Institute, Durham, North Carolina
| | - Gerard Socie
- Hematology Transplantation, AP-HP Saint Louis Hospital & University of Paris, INSERM U976, Paris, France
| | - Hildegard Greinix
- Clinical Division of Hematology, Medical University of Graz, Graz, Austria
| | - Corey Cutler
- Division of Stem Cell Transplantation and Cellular Therapy, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
24
|
Artificial Intelligence in Clinical Immunology. Artif Intell Med 2021. [DOI: 10.1007/978-3-030-58080-3_83-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
Eckardt JN, Bornhäuser M, Wendt K, Middeke JM. Application of machine learning in the management of acute myeloid leukemia: current practice and future prospects. Blood Adv 2020; 4:6077-6085. [PMID: 33290546 PMCID: PMC7724910 DOI: 10.1182/bloodadvances.2020002997] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Machine learning (ML) is rapidly emerging in several fields of cancer research. ML algorithms can deal with vast amounts of medical data and provide a better understanding of malignant disease. Its ability to process information from different diagnostic modalities and functions to predict prognosis and suggest therapeutic strategies indicates that ML is a promising tool for the future management of hematologic malignancies; acute myeloid leukemia (AML) is a model disease of various recent studies. An integration of these ML techniques into various applications in AML management can assure fast and accurate diagnosis as well as precise risk stratification and optimal therapy. Nevertheless, these techniques come with various pitfalls and need a strict regulatory framework to ensure safe use of ML. This comprehensive review highlights and discusses recent advances in ML techniques in the management of AML as a model disease of hematologic neoplasms, enabling researchers and clinicians alike to critically evaluate this upcoming, potentially practice-changing technology.
Collapse
Affiliation(s)
- Jan-Niklas Eckardt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- National Center for Tumor Diseases, Dresden (NCT/UCC), Dresden, Germany
- German Consortium for Translational Cancer Research, DKFZ, Heidelberg, Germany; and
| | - Karsten Wendt
- Institute of Circuits and Systems, Technical University Dresden, Dresden, Germany
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
26
|
Barone SM, Paul AG, Muehling LM, Lannigan JA, Kwok WW, Turner RB, Woodfolk JA, Irish JM. Unsupervised machine learning reveals key immune cell subsets in COVID-19, rhinovirus infection, and cancer therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.31.190454. [PMID: 32766581 PMCID: PMC7402038 DOI: 10.1101/2020.07.31.190454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For an emerging disease like COVID-19, systems immunology tools may quickly identify and quantitatively characterize cells associated with disease progression or clinical response. With repeated sampling, immune monitoring creates a real-time portrait of the cells reacting to a novel virus before disease specific knowledge and tools are established. However, single cell analysis tools can struggle to reveal rare cells that are under 0.1% of the population. Here, the machine learning workflow Tracking Responders Expanding (T-REX) was created to identify changes in both very rare and common cells in diverse human immune monitoring settings. T-REX identified cells that were highly similar in phenotype and localized to hotspots of significant change during rhinovirus and SARS-CoV-2 infections. Specialized reagents used to detect the rhinovirus-specific CD4+ cells, MHCII tetramers, were not used during unsupervised analysis and instead 'left out' to serve as a test of whether T-REX identified biologically significant cells. In the rhinovirus challenge study, T-REX identified virus-specific CD4+ T cells based on these cells being a distinct phenotype that expanded by ≥95% following infection. T-REX successfully identified hotspots containing virus-specific T cells using pairs of samples comparing Day 7 of infection to samples taken either prior to infection (Day 0) or after clearing the infection (Day 28). Mapping pairwise comparisons in samples according to both the direction and degree of change provided a framework to compare systems level immune changes during infectious disease or therapy response. This revealed that the magnitude and direction of systemic immune change in some COVID-19 patients was comparable to that of blast crisis acute myeloid leukemia patients undergoing induction chemotherapy and characterized the identity of the immune cells that changed the most. Other COVID-19 patients instead matched an immune trajectory like that of individuals with rhinovirus infection or melanoma patients receiving checkpoint inhibitor therapy. T-REX analysis of paired blood samples provides an approach to rapidly identify and characterize mechanistically significant cells and to place emerging diseases into a systems immunology context.
Collapse
Affiliation(s)
- Sierra M. Barone
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberta G.A. Paul
- Allergy Division, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lyndsey M. Muehling
- Allergy Division, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Joanne A. Lannigan
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - William W. Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Ronald B. Turner
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Judith A. Woodfolk
- Allergy Division, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jonathan M. Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
27
|
Gupta V, Braun TM, Chowdhury M, Tewari M, Choi SW. A Systematic Review of Machine Learning Techniques in Hematopoietic Stem Cell Transplantation (HSCT). SENSORS (BASEL, SWITZERLAND) 2020; 20:E6100. [PMID: 33120974 PMCID: PMC7663237 DOI: 10.3390/s20216100] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 12/11/2022]
Abstract
Machine learning techniques are widely used nowadays in the healthcare domain for the diagnosis, prognosis, and treatment of diseases. These techniques have applications in the field of hematopoietic cell transplantation (HCT), which is a potentially curative therapy for hematological malignancies. Herein, a systematic review of the application of machine learning (ML) techniques in the HCT setting was conducted. We examined the type of data streams included, specific ML techniques used, and type of clinical outcomes measured. A systematic review of English articles using PubMed, Scopus, Web of Science, and IEEE Xplore databases was performed. Search terms included "hematopoietic cell transplantation (HCT)," "autologous HCT," "allogeneic HCT," "machine learning," and "artificial intelligence." Only full-text studies reported between January 2015 and July 2020 were included. Data were extracted by two authors using predefined data fields. Following PRISMA guidelines, a total of 242 studies were identified, of which 27 studies met the inclusion criteria. These studies were sub-categorized into three broad topics and the type of ML techniques used included ensemble learning (63%), regression (44%), Bayesian learning (30%), and support vector machine (30%). The majority of studies examined models to predict HCT outcomes (e.g., survival, relapse, graft-versus-host disease). Clinical and genetic data were the most commonly used predictors in the modeling process. Overall, this review provided a systematic review of ML techniques applied in the context of HCT. The evidence is not sufficiently robust to determine the optimal ML technique to use in the HCT setting and/or what minimal data variables are required.
Collapse
Affiliation(s)
- Vibhuti Gupta
- Michigan Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas M. Braun
- School of Public Health, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Mosharaf Chowdhury
- Michigan Engineering, Computer Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Muneesh Tewari
- Michigan Medicine, Department of Internal Medicine, Hematology/Oncology Division, University of Michigan, Ann Arbor, MI 48109, USA;
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Engineering, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sung Won Choi
- Michigan Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
28
|
|
29
|
Radakovich N, Cortese M, Nazha A. Acute myeloid leukemia and artificial intelligence, algorithms and new scores. Best Pract Res Clin Haematol 2020; 33:101192. [PMID: 33038981 PMCID: PMC7548395 DOI: 10.1016/j.beha.2020.101192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/27/2020] [Indexed: 12/23/2022]
Abstract
Artificial intelligence, and more narrowly machine-learning, is beginning to expand humanity's capacity to analyze increasingly large and complex datasets. Advances in computer hardware and software have led to breakthroughs in multiple sectors of our society, including a burgeoning role in medical research and clinical practice. As the volume of medical data grows at an apparently exponential rate, particularly since the human genome project laid the foundation for modern genetic inquiry, informatics tools like machine learning are becoming crucial in analyzing these data to provide meaningful tools for diagnostic, prognostic, and therapeutic purposes. Within medicine, hematologic diseases can be particularly challenging to understand and treat given the increasingly complex and intercalated genetic, epigenetic, immunologic, and regulatory pathways that must be understood to optimize patient outcomes. In acute myeloid leukemia (AML), new developments in machine learning algorithms have enabled a deeper understanding of disease biology and the development of better prognostic and predictive tools. Ongoing work in the field brings these developments incrementally closer to clinical implementation.
Collapse
Affiliation(s)
- Nathan Radakovich
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, United States
| | - Matthew Cortese
- Department of Hematology and Medical Oncology, Cleveland Clinic, United States
| | - Aziz Nazha
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, United States; Department of Hematology and Medical Oncology, Cleveland Clinic, United States; Center for Clinical Artificial Intelligence, Cleveland Clinic, United States.
| |
Collapse
|
30
|
Machine learning for predicting long-term kidney allograft survival: a scoping review. Ir J Med Sci 2020; 190:807-817. [PMID: 32761550 DOI: 10.1007/s11845-020-02332-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/26/2020] [Indexed: 12/24/2022]
Abstract
Supervised machine learning (ML) is a class of algorithms that "learn" from existing input-output pairs, which is gaining popularity in pattern recognition for classification and prediction problems. In this scoping review, we examined the use of supervised ML algorithms for the prediction of long-term allograft survival in kidney transplant recipients. Data sources included PubMed, the Cumulative Index to Nursing and Allied Health Literature, and the Institute for Electrical and Electronics Engineers (IEEE) Xplore libraries from inception to November 2019. We screened titles and abstracts and potentially eligible full-text reports to select studies and subsequently abstracted the data. Eleven studies were identified. Decision trees were the most commonly used method (n = 8), followed by artificial neural networks (ANN) (n = 4) and Bayesian belief networks (n = 2). The area under receiver operating curve (AUC) was the most common measure of discrimination (n = 7), followed by sensitivity (n = 5) and specificity (n = 4). Model calibration examining the reliability in risk prediction was performed using either the Pearson r or the Hosmer-Lemeshow test in four studies. One study showed that logistic regression had comparable performance to ANN, while another study demonstrated that ANN performed better in terms of sensitivity, specificity, and accuracy, as compared with a Cox proportional hazards model. We synthesized the evidence related to the comparison of ML techniques with traditional statistical approaches for prediction of long-term allograft survival in patients with a kidney transplant. The methodological and reporting quality of included studies was poor. Our study also demonstrated mixed results in terms of the predictive potential of the models.
Collapse
|
31
|
Machine learning in haematological malignancies. LANCET HAEMATOLOGY 2020; 7:e541-e550. [PMID: 32589980 DOI: 10.1016/s2352-3026(20)30121-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/02/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Machine learning is a branch of computer science and statistics that generates predictive or descriptive models by learning from training data rather than by being rigidly programmed. It has attracted substantial attention for its many applications in medicine, both as a catalyst for research and as a means of improving clinical care across the cycle of diagnosis, prognosis, and treatment of disease. These applications include the management of haematological malignancy, in which machine learning has created inroads in pathology, radiology, genomics, and the analysis of electronic health record data. As computational power becomes cheaper and the tools for implementing machine learning become increasingly democratised, it is likely to become increasingly integrated into the research and practice landscape of haematology. As such, machine learning merits understanding and attention from researchers and clinicians alike. This narrative Review describes important concepts in machine learning for unfamiliar readers, details machine learning's current applications in haematological malignancy, and summarises important concepts for clinicians to be aware of when appraising research that uses machine learning.
Collapse
|
32
|
Shouval R, Fein JA, Savani B, Mohty M, Nagler A. Machine learning and artificial intelligence in haematology. Br J Haematol 2020; 192:239-250. [PMID: 32602593 DOI: 10.1111/bjh.16915] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Digitalization of the medical record and integration of genomic methods into clinical practice have resulted in an unprecedented wealth of data. Machine learning is a subdomain of artificial intelligence that attempts to computationally extract meaningful insights from complex data structures. Applications of machine learning in haematological scenarios are steadily increasing. However, basic concepts are often unfamiliar to clinicians and investigators. The purpose of this review is to provide readers with tools to interpret and critically appraise machine learning literature. We begin with the elucidation of standard terminology and then review examples in haematology. Guidelines for designing and evaluating machine-learning studies are provided. Finally, we discuss limitations of the machine-learning approach.
Collapse
Affiliation(s)
- Roni Shouval
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Hematology and Bone Marrow Transplantation Division, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Joshua A Fein
- University of Connecticut Medical Center, Farmington, CT, USA
| | - Bipin Savani
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohamad Mohty
- European Society for Blood and Marrow Transplantation Paris Study Office/CEREST-TC, Paris, France.,Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Arnon Nagler
- Hematology and Bone Marrow Transplantation Division, Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
33
|
Leelatian N, Sinnaeve J, Mistry AM, Barone SM, Brockman AA, Diggins KE, Greenplate AR, Weaver KD, Thompson RC, Chambless LB, Mobley BC, Ihrie RA, Irish JM. Unsupervised machine learning reveals risk stratifying glioblastoma tumor cells. eLife 2020; 9:56879. [PMID: 32573435 PMCID: PMC7340505 DOI: 10.7554/elife.56879] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
A goal of cancer research is to reveal cell subsets linked to continuous clinical outcomes to generate new therapeutic and biomarker hypotheses. We introduce a machine learning algorithm, Risk Assessment Population IDentification (RAPID), that is unsupervised and automated, identifies phenotypically distinct cell populations, and determines whether these populations stratify patient survival. With a pilot mass cytometry dataset of 2 million cells from 28 glioblastomas, RAPID identified tumor cells whose abundance independently and continuously stratified patient survival. Statistical validation within the workflow included repeated runs of stochastic steps and cell subsampling. Biological validation used an orthogonal platform, immunohistochemistry, and a larger cohort of 73 glioblastoma patients to confirm the findings from the pilot cohort. RAPID was also validated to find known risk stratifying cells and features using published data from blood cancer. Thus, RAPID provides an automated, unsupervised approach for finding statistically and biologically significant cells using cytometry data from patient samples.
Collapse
Affiliation(s)
- Nalin Leelatian
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Justine Sinnaeve
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States
| | - Akshitkumar M Mistry
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Sierra M Barone
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Asa A Brockman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States
| | - Kirsten E Diggins
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States
| | - Allison R Greenplate
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Kyle D Weaver
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Reid C Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Lola B Chambless
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, United States
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| |
Collapse
|
34
|
Adom D, Rowan C, Adeniyan T, Yang J, Paczesny S. Biomarkers for Allogeneic HCT Outcomes. Front Immunol 2020; 11:673. [PMID: 32373125 PMCID: PMC7186420 DOI: 10.3389/fimmu.2020.00673] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 03/25/2020] [Indexed: 12/23/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (HCT) remains the only curative therapy for many hematological malignant and non-malignant disorders. However, key obstacles to the success of HCT include graft-versus-host disease (GVHD) and disease relapse due to absence of graft-versus-tumor (GVT) effect. Over the last decade, advances in "omics" technologies and systems biology analysis, have allowed for the discovery and validation of blood biomarkers that can be used as diagnostic test and prognostic test (that risk-stratify patients before disease occurrence) for acute and chronic GVHD and recently GVT. There are also predictive biomarkers that categorize patients based on their likely to respond to therapy. Newer mathematical analysis such as machine learning is able to identify different predictors of GVHD using clinical characteristics pre-transplant and possibly in the future combined with other biomarkers. Biomarkers are not only useful to identify patients with higher risk of disease progression, but also help guide treatment decisions and/or provide a basis for specific therapeutic interventions. This review summarizes biomarkers definition, omics technologies, acute, chronic GVHD and GVT biomarkers currently used in clinic or with potential as targets for existing or new drugs focusing on novel published work.
Collapse
Affiliation(s)
- Djamilatou Adom
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Courtney Rowan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Titilayo Adeniyan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jinfeng Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
35
|
Serrano-López J, Fernández JL, Lumbreras E, Serrano J, Martínez-Losada C, Martín C, Hernández-Rivas JM, Sánchez-García J. Machine learning applied to gene expression analysis of T-lymphocytes in patients with cGVHD. Bone Marrow Transplant 2020; 55:1668-1670. [PMID: 32157244 DOI: 10.1038/s41409-020-0848-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/18/2020] [Accepted: 02/25/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Juana Serrano-López
- Instituto Maimonides Investigación Biomédica, IMIBIC/University of Córdoba, Córdoba, Spain. .,Experimental Hematology Lab, IIS-Fundación Jiménez Díaz, UAM, Madrid, Spain.
| | - José Luis Fernández
- Instituto Maimonides Investigación Biomédica, IMIBIC/University of Córdoba, Córdoba, Spain
| | - Eva Lumbreras
- Centro de Investigación del Cancer, Universidad de Salamanca, CSIC, Salamanca, Spain
| | - Josefina Serrano
- Instituto Maimonides Investigación Biomédica, IMIBIC/University of Córdoba, Córdoba, Spain.,Hematology Department, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Carmen Martínez-Losada
- Instituto Maimonides Investigación Biomédica, IMIBIC/University of Córdoba, Córdoba, Spain.,Hematology Department, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Carmen Martín
- Instituto Maimonides Investigación Biomédica, IMIBIC/University of Córdoba, Córdoba, Spain.,Hematology Department, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Jesús M Hernández-Rivas
- Centro de Investigación del Cancer, Universidad de Salamanca, CSIC, Salamanca, Spain.,Hospital Universitario de Salamanca IBSAL, Salamanca, Spain
| | - Joaquín Sánchez-García
- Instituto Maimonides Investigación Biomédica, IMIBIC/University of Córdoba, Córdoba, Spain.,Hematology Department, Hospital Universitario Reina Sofía, Córdoba, Spain
| |
Collapse
|
36
|
Martin PJ, Storer BE, Palmer J, Jagasia MH, Chen GL, Broady R, Arora M, Pidala JA, Hamilton BK, Lee SJ. Organ Changes Associated with Provider-Assessed Responses in Patients with Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2019; 25:1869-1874. [PMID: 31085305 PMCID: PMC6755054 DOI: 10.1016/j.bbmt.2019.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 12/21/2022]
Abstract
Assessments of overall improvement and worsening of chronic graft-versus-host disease (GVHD) manifestations by the algorithm recommended by National Institutes of Health (NIH) response criteria do not align closely with those reported by providers, particularly when patients have mixed responses with improvement in some manifestations but worsening in others. To elucidate the changes that influence provider assessment of response, we used logistic regression to generate an overall change index based on specific manifestations of chronic GVHD measured at baseline and 6 months later. We hypothesized that this overall change index would correlate strongly with overall improvement as determined by providers. The analysis included 488 patients from 2 prospective observational studies who were randomly assigned in a 3:2 ratio to discovery and replication cohorts. Changes in bilirubin and scores of the lower gastrointestinal tract, mouth, joint/fascia, lung, and skin were correlated with provider-assessed improvement, suggesting that the main NIH response measures capture relevant information. Conversely, changes in the eye, esophagus, and upper gastrointestinal tract did not correlate with provider-assessed response, suggesting that these scales could be modified or dropped from the NIH response assessment. The area under the receiver operator characteristic curve in the replication cohort was 0.72, indicating that the scoring algorithm for overall change based on NIH response measures is not well calibrated with provider-assessed response.
Collapse
Affiliation(s)
- Paul J Martin
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington.
| | - Barry E Storer
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Madan H Jagasia
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - George L Chen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Raewyn Broady
- Leukemia/Bone Marrow Transplant Program of British Columbia, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Mukta Arora
- Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Joseph A Pidala
- Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Betty K Hamilton
- Blood and Marrow Transplantation, Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
37
|
Wolff D, Hilgendorf I, Wagner-Drouet E, Jedlickova Z, Ayuk F, Zeiser R, Schäfer-Eckart K, Gerbitz A, Stadler M, Klein S, Middeke JM, Lawitschka A, Winkler J, Halter J, Holler E, Kobbe G, Stelljes M, Ditschkowski M, Greinix H. Changes in Immunosuppressive Treatment of Chronic Graft-versus-Host Disease: Comparison of 2 Surveys within Allogeneic Hematopoietic Stem Cell Transplant Centers in Germany, Austria, and Switzerland. Biol Blood Marrow Transplant 2019; 25:1450-1455. [PMID: 30876928 DOI: 10.1016/j.bbmt.2019.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/04/2019] [Indexed: 12/27/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) remains the leading cause of late morbidity and mortality. Despite the growing number of treatment options in cGVHD, evidence remains sparse. The German-Austrian-Swiss GVHD Consortium performed a survey on clinical practice in treatment of cGVHD among transplant centers in Germany, Austria, and Switzerland in 2009 and 2018 and compared the results. The survey performed in 2009 contained 20 questions on first-line treatment and related issues and 4 questions on second-line scenarios followed by a survey on all systemic and topic treatment options known and applied, with 31 of 36 transplant centers (86%) responding. The survey in 2018 repeated 7 questions on first-line treatment and 3 questions on second-line scenarios followed by an updated survey on all current systemic treatment options known and applied, with 29 of 66 centers (43%) responding. In summary, the results show a large overlap of first-line treatment practice between centers and the 2 surveys because of a lack of new data that changes practice, except significant heterogeneity of treatment of cGVHD progressive onset type, which can be explained by the lack of trials focusing on this high-risk entity. In contrast, treatment options applied to second-line therapy vary considerably, with new agents like ibrutinib and ruxolitinib entering clinical practice. Moreover, treatment of bronchiolitis obliterans syndrome demonstrates heterogeneity in applied therapeutic options and sequence because of a lack of controlled data and different conclusions from already existing evidence. In summary, the survey results demonstrate an increasing number of treatment options applied to cGVHD accompanied by a significant heterogeneity in second-line treatment and underline the urgent need for clinical trials and registry analyses on rare entities with high mortality like progressive onset type and lung involvement of cGVHD.
Collapse
Affiliation(s)
- Daniel Wolff
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.
| | - Inken Hilgendorf
- Klinik für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Jena, Germany
| | - Eva Wagner-Drouet
- 3rd Medical Department, Hematology, Oncology and Pneumology, University Medical Center, Mainz, Germany
| | - Zuzana Jedlickova
- Medizinische Klinik 2, Universitätsklinikum Frankfurt, Frankfurt/Main, Germany
| | - Francis Ayuk
- Department of Stem Cell Transplantation, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, University of Freiburg, Freiburg, Germany
| | | | - Armin Gerbitz
- Department of Hematology, Oncology and Tumorimmunology, Campus Virchow Klinikum, Charite, Berlin, Germany
| | - Michael Stadler
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Stefan Klein
- III. Medizinische Klinik Hämatologie und Onkologie, Universitätsmedizin Mannheim, Mannheim, Germany
| | - Jan-Moritz Middeke
- Department of Internal Medicine I, University Hospital Dresden, Dresden, Germany
| | - Anita Lawitschka
- St. Anna Children's Hospital, Medical University Vienna, Austria
| | - Julia Winkler
- Department of Hematology, University Hospital Erlangen, Erlangen, Germany
| | - Jörg Halter
- Department of Hematology, University Hospital Basel, Basel, Switzerland
| | - Ernst Holler
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Matthias Stelljes
- Department of Hematology, University Hospital Muenster, Muenster, Germany
| | - Markus Ditschkowski
- Department for Bone Marrow Transplantation, University of Essen, Essen, Germany
| | - Hildegard Greinix
- Division of Hematology, Department of Internal Medicine I, Medical University of Graz, Graz, Austria
| |
Collapse
|