1
|
Parker RS, Le J, Villa M, Luong A, Lin TY, Lee Y, Doan A, Aguayo-Hiraldo P, Pannaraj PS, Yoon SJ, Wallace WD, Armstrong A, O’Gorman MR, Bard JD, Parekh C. COVID-19 vaccinated children, adolescents, and young adults with acute lymphoblastic leukemia show spike reactive antibodies and multifunctional T-cells. Int J Cancer 2024; 155:2190-2200. [PMID: 39005114 PMCID: PMC11499007 DOI: 10.1002/ijc.35096] [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: 12/06/2023] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024]
Abstract
Little is known about the efficacy of COVID-19 vaccines during acute lymphoblastic leukemia therapy (ALL); data for COVID-19 vaccine immune responses in pediatric leukemia remain sparse. We conducted a single center study of patients aged 5-25 years undergoing ALL chemotherapy who received COVID-19 vaccination. Twenty-one patients were enrolled; efficacy was evaluable in 20. Twenty were vaccinated while receiving chemotherapy. Twenty received the BNT162b2 mRNA vaccine. Spike reactive antibodies (S-IgG) and/or T-cells (SRT) were detected in 16 of 20 (80%) vaccinated patients; 13 (65%) and 9 (45%) were positive for S-IgG and SRT, respectively. Six (30%) showed both spike reactive B and T-cell responses. Eleven of the 13 with S-IgG positivity were negative for anti-Nucleocapsid IgG, an antibody profile consistent with a vaccine induced immune response. All 13S-IgG+ patients showed neutralizing antibodies. SRT included CD4+ (7) and CD8+ (6) T-cells; both CD4+ and CD8+ SRT were seen in 4. SRT were multifunctional (producing multiple cytokines) in most patients (8 of 9); 4 showed SRT with triple cytokine and B-cell co-stimulatory responses, indicating a multimodal adaptive immune response. Immune responses were seen among patients vaccinated in the settings of lymphopenia (6 of 12) intensive chemotherapy (3 of 4), and Peg allergy (6 of 8). Sequencing revealed public CD4+ and CD8+ TCR sequences reactive to epitopes across the spike protein. In conclusion, COVID-19 vaccination induced B and/or T-cell responses in a majority of children and young adults undergoing ALL chemotherapy.
Collapse
Affiliation(s)
- Rebecca S Parker
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Justin Le
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Miguel Villa
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Annie Luong
- The Saban Research institute, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Tsen Yin Lin
- The Saban Research institute, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Yesun Lee
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, University of California San Diego, San Diego, California, USA
| | - Andrew Doan
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Paibel Aguayo-Hiraldo
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Pia S Pannaraj
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, University of California San Diego, San Diego, California, USA
| | - Seon-Jae Yoon
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - William Dean Wallace
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - April Armstrong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Maurice R O’Gorman
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Jennifer Dien Bard
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Chintan Parekh
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
2
|
Sherman AC, Tuan J, Cantos VD, Adeyiga O, Mahoney S, Ortega-Villa AM, Tillman A, Whitaker J, Woodward Davis AS, Leav B, Hirsch I, Sadoff J, Dunkle LM, Gilbert PB, Janes HE, Kublin JG, Goepfert PA, Kotloff K, Rouphael N, Falsey AR, El Sahly HM, Sobieszczyk ME, Huang Y, Neuzil KM, Corey L, Grinsztejn B, Gray G, Nason M, Baden LR, Gay CL. COVID-19 Vaccine Efficacy in Participants With Weakened Immune Systems From 4 Randomized Controlled Trials. Clin Infect Dis 2024; 79:364-374. [PMID: 38598658 PMCID: PMC11327800 DOI: 10.1093/cid/ciae192] [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: 12/28/2023] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Although the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are highly efficacious at preventing severe disease in the general population, current data are lacking regarding vaccine efficacy (VE) for individuals with mild immunocompromising conditions. METHODS A post hoc, cross-protocol analysis of participant-level data from the blinded phase of four randomized, placebo-controlled, coronavirus disease 2019 (COVID-19) vaccine phase 3 trials (Moderna, AstraZeneca, Janssen, and Novavax) was performed. We defined a "tempered immune system" (TIS) variable via a consensus panel based on medical history and medications to determine VE against symptomatic and severe COVID-19 cases in TIS participants versus non-TIS individuals starting at 14 days after completion of the primary series through the blinded phase for each of the 4 trials. An analysis of participants living with well-controlled human immunodeficiency virus was conducted using the same methods. RESULTS A total of 3852/30 351 (12.7%) Moderna participants, 3088/29 868 (10.3%) Novavax participants, 3549/32 380 (11.0%) AstraZeneca participants, and 5047/43 788 (11.5%) Janssen participants were identified as having a TIS. Most TIS conditions (73.9%) were due to metabolism and nutritional disorders. Vaccination (vs placebo) significantly reduced the likelihood of symptomatic and severe COVID-19 for all participants for each trial. VE was not significantly different for TIS participants versus non-TIS for either symptomatic or severe COVID-19 for each trial, nor was VE significantly different in the symptomatic endpoint for participants with human immunodeficiency virus. CONCLUSIONS For individuals with mildly immunocompromising conditions, there is no evidence of differences in VE against symptomatic or severe COVID-19 compared with those with non-TIS in the 4 COVID-19 vaccine randomized controlled efficacy trials.
Collapse
Affiliation(s)
- Amy C Sherman
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica Tuan
- Yale School of Medicine, Section of Infectious Diseases, New Haven, Connecticut, USA
| | - Valeria D Cantos
- Division of Infectious Diseases, Emory University, Atlanta, Georgia, USA
| | - Oladunni Adeyiga
- Department of Medicine, Division of Infectious Diseases, University of California, Los Angeles, Los Angeles, California, USA
| | - Scott Mahoney
- Department of Medicine, University of Cape Town, Desmond Tutu HIV Centre, Cape Town, South Africa
| | - Ana M Ortega-Villa
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Amy Tillman
- Frederick National Laboratory for Cancer Research, Clinical Monitoring Research Program Directorate, Frederick, Maryland, USA
| | - Jennifer Whitaker
- Department of Molecular Virology and Microbiology and Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Amanda S Woodward Davis
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Brett Leav
- Moderna Inc., Cambridge, Massachusetts, USA
| | - Ian Hirsch
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | | | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Holly E Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Karen Kotloff
- Department of Pediatrics and the Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Ann R Falsey
- Infectious Disease Division, University of Rochester, Rochester, New York, USA
| | - Hana M El Sahly
- Department of Molecular Virology and Microbiology and Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | | | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Beatriz Grinsztejn
- National Institute of Infectious Diseases-Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Glenda Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Martha Nason
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lindsey R Baden
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Cynthia L Gay
- Department of Medicine, Division of Infectious Diseases, UNC HIV Cure Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
Gai X, Sun X, Liu B, Yan W, Sheng Z, Zhou Q, Sun Y. Efficacy of Combination of Antiviral Therapy With Neutralizing Monoclonal Antibodies for Recurrent Persistent SARS-CoV-2 Pneumonia in Patients With Lymphoma. BIOMED RESEARCH INTERNATIONAL 2024; 2024:8182887. [PMID: 39140001 PMCID: PMC11321881 DOI: 10.1155/2024/8182887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 08/15/2024]
Abstract
Despite the potential of neutralizing antibodies in the management of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), clinical research on its efficacy in Chinese patients remains limited. This study is aimed at investigating the therapeutic effect of combination of antiviral therapy with neutralizing monoclonal antibodies for recurrent persistent SARS-CoV-2 pneumonia in patients with lymphoma complicated by B cell depletion. A prospective study was conducted on Chinese patients who were treated with antiviral nirmatrelvir/ritonavir therapy and the neutralizing antibody tixagevimab-cilgavimab (tix-cil). The primary outcome was the rate of recurrent SARS-CoV-2 infection. Five patients with lymphoma experienced recurrent SARS-CoV-2 pneumonia and received tix-cil treatment. All patients had a history of CD20 monoclonal antibody use within the year preceding SARS-CoV-2 infection, and two patients also had a history of Bruton's tyrosine kinase (BTK) inhibitor use. These patients had notably low lymphocyte counts and exhibited near depletion of B cells. All five patients tested negative for serum SARS-CoV-2 IgG and IgM antibodies. None of the patients developed reinfection with SARS-CoV-2 pneumonia after antiviral and tix-cil treatment during the 6-month follow-up period. In conclusion, the administration of antiviral and SARS-CoV-2-neutralizing antibodies showed encouraging therapeutic efficacy against SARS-CoV-2 pneumonia in patients with lymphoma complicated by B cell depletion, along with the potential preventive effect of neutralizing antibodies for up to 6 months.
Collapse
Affiliation(s)
- Xiaoyan Gai
- Department of Pulmonary and Critical Care Medicine Peking University Third Hospital, Beijing 100083, China
| | - Xiaoyan Sun
- Department of Pulmonary and Critical Care Medicine Peking University Third Hospital, Beijing 100083, China
| | - Beibei Liu
- Department of Pulmonary and Critical Care Medicine Peking University Third Hospital, Beijing 100083, China
| | - Wei Yan
- Department of Pulmonary and Critical Care Medicine Peking University Third Hospital, Beijing 100083, China
| | - Zikang Sheng
- Department of Pulmonary and Critical Care Medicine Peking University Third Hospital, Beijing 100083, China
| | - Qingtao Zhou
- Department of Pulmonary and Critical Care Medicine Peking University Third Hospital, Beijing 100083, China
| | - Yongchang Sun
- Department of Pulmonary and Critical Care Medicine Peking University Third Hospital, Beijing 100083, China
| |
Collapse
|
4
|
Moreno Echevarria F, Caputo M, Camp D, Reddy S, Achenbach CJ. Incidence and risk factors of SARS-CoV-2 breakthrough infection in the early Omicron variant era among vaccinated and boosted individuals in Chicago. PLoS One 2024; 19:e0302338. [PMID: 39102410 PMCID: PMC11299831 DOI: 10.1371/journal.pone.0302338] [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: 04/01/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND SARS-CoV-2 vaccines are safe and effective against infection and severe COVID-19 disease worldwide. Certain co-morbid conditions cause immune dysfunction and may reduce immune response to vaccination. In contrast, those with co-morbidities may practice infection prevention strategies. Thus, the real-world clinical impact of co-morbidities on SARS-CoV-2 infection in the recent post-vaccination period is not well established. This study was performed to understand the epidemiology of Omicron breakthrough infection and evaluate associations with number of comorbidities in a vaccinated and boosted population. METHODS AND FINDINGS A retrospective clinical cohort study was performed utilizing the Northwestern Medicine Enterprise Data Warehouse. Our study population was identified as fully vaccinated adults with at least one booster. The primary risk factor of interest was the number of co-morbidities. The primary outcome was the incidence and time to the first positive SARS-CoV-2 molecular test in the Omicron predominant era. Multivariable Cox modeling analyses to determine the hazard of SARS-CoV-2 infection were stratified by calendar time (Period 1: January 1 -June 30, 2022; Period 2: July 1 -December 31, 2022) due to violations in the proportional hazards assumption. In total, 133,191 patients were analyzed. During Period 1, having 3+ comorbidities was associated with increased hazard for breakthrough (HR = 1.16 CI 1.08-1.26). During Period 2 of the study, having 2 comorbidities (HR = 1.45 95% CI 1.26-1.67) and having 3+ comorbidities (HR 1.73, 95% CI 1.51-1.97) were associated with increased hazard for Omicron breakthrough. Older age was associated with decreased hazard in Period 1 of follow-up. Interaction terms for calendar time indicated significant changes in hazard for many factors between the first and second halves of the follow-up period. CONCLUSIONS Omicron breakthrough is common with significantly higher risk for our most vulnerable patients with multiple co-morbidities. Age plays an important role in breakthrough infection with the highest incidence among young adults, which may be due to age-related behavioral factors. These findings reflect real-world differences in immunity and exposure risk behaviors for populations vulnerable to COVID-19.
Collapse
Affiliation(s)
- Fabiola Moreno Echevarria
- Robert J. Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Mathew Caputo
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Daniel Camp
- Robert J. Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Susheel Reddy
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Chad J. Achenbach
- Robert J. Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Chicago, Illinois, United States of America
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
5
|
Yang S, Wei R, Shi H, Wang Y, Lai Y, Zhao X, Lu J, Schmitz N. The impact of Bruton's tyrosine kinase inhibitor treatment on COVID-19 outcomes in Chinese patients with chronic lymphocytic leukemia. Front Oncol 2024; 14:1396913. [PMID: 38835372 PMCID: PMC11148257 DOI: 10.3389/fonc.2024.1396913] [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: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024] Open
Abstract
Background Impact of B-cell depletion following treatment with Bruton tyrosine kinase-inhibitors (BTKi) on the outcome of SARS-CoV-2 infection in chronic lymphocytic leukemia (CLL) patients remain controversial. We investigated the impact of BTKi on susceptibility and the severity of COVID-19 in Chinese patients with CLL during the first wave of COVID-19 (Omicron variant). Methods CLL patients (n=171) visiting the Institute of Hematology, Peoples' Hospital, China (November 15, 2022- January 20, 2023) were included in the study. Seventeen patients receiving BTKi and venetoclax with or without obinutuzumab were excluded. Data from 117 patients receiving treatment with BTKi were collected using a standardized questionnaire through telephone interviews. Thirty-four patients without CLL-specific treatment served as controls. The data was analysed using IBM SPSS Software version 21 and a P value of <0.05 was considered statistically significant. Results The median age of patients was 67 years and majority were males (n=100). Treatment with BTKi was not associated with higher incidence of COVID-19 (74% [95% Confidence Interval (CI) 60%, 92%]) versus 74% (CI 48%, 100%) without any treatment (P=0.92). Hypoxemia was reported by 45% (32%, 61%) and 16% (4%, 41%) (P=0.01). BTKi was the only independent risk factor of hypoxemia (Hazard Ratio [HR], 4.22 [1.32, 13.50]; P = 0.02). Five (5.7%) patients with COVID-19 under BTKi required ICU admission; 4 of them died. No ICU admissions/deaths were observed in the control group. Conclusion In Chinese patients with CLL and treated with BTKi experienced more severe lung disease and ICU admissions due to COVID-19 than patients without CLL therapy. Frequency of infections with SARS-CoV-2, however, was not different in patients with or without BTKi treatment.
Collapse
Affiliation(s)
- Shenmiao Yang
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
| | - Rong Wei
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
| | - Hongxia Shi
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
| | - Yazhe Wang
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
| | - Yueyun Lai
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
| | - Xiaosu Zhao
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
| | - Jin Lu
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
| | - Norbert Schmitz
- Peking University Institute of Hematology, Peking University Peoples' Hospital, Beijing, China
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
6
|
Harandi H, Fallahtafti P, Karimi A, Hashemi SM, Mahalleh M, Ashouri M, Salehi MA, Hoveidaei A. Examining the immunological responses to COVID-19 vaccination in multiple myeloma patients: a systematic review and meta-analysis. BMC Geriatr 2024; 24:411. [PMID: 38720296 PMCID: PMC11080142 DOI: 10.1186/s12877-024-05006-0] [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: 02/11/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Impaired immune response in multiple myeloma renders the patients vulnerable to infections, such as COVID-19, and may cause worse response to vaccines. Researchers should analyze this issue to enable the planning for special preventive measures, such as increased booster doses. Therefore, this meta-analysis aimed to evaluate the response and efficacy of COVID-19 vaccines in patients with multiple myeloma. METHODS This meta-analysis followed PRISMA 2020 guidelines, conducting a comprehensive database search using specified keywords. Study selection involved a two-phase title/abstract and full-text screening process. Data extraction was performed by two researchers, and statistical analysis involved meta-analysis, subgroup analysis based on vaccine dosage and study time, random effects meta-regression, and heterogeneity testing using the Q test. RESULTS The meta-analysis revealed that patients with multiple myeloma (MM) had a lower likelihood of developing detectable antibodies after COVID-19 vaccination compared to healthy controls (Log odds ratio with 95% CI: -3.34 [-4.08, -2.60]). The analysis of antibody response after different doses showed consistent lower seropositivity in MM patients (after first dose: -2.09, [-3.49, -0.69], second: -3.80, 95%CI [-4.71, -3.01], a booster dose: -3.03, [-5.91, -0.15]). However, there was no significant difference in the mean level of anti-S antibodies between MM patients and controls (Cohen's d -0.72, [-1.86, 0.43]). Evaluation of T-cell responses indicated diminished T-cell-mediated immunity in MM patients compared to controls. Seven studies reported clinical response, with breakthrough infections observed in vaccinated MM patients. CONCLUSIONS These findings highlight the impaired humoral and cellular immune responses in MM patients after COVID-19 vaccination, suggesting the need for further investigation and potential interventions.
Collapse
Affiliation(s)
- Hamid Harandi
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Fallahtafti
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Uro-Oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | | - Mehrdad Mahalleh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Moein Ashouri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Armin Hoveidaei
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Merli M, Costantini A, Tafuri S, Bavaro DF, Minoia C, Meli E, Luminari S, Gini G. Management of vaccinations in patients with non-Hodgkin lymphoma. Br J Haematol 2024; 204:1617-1634. [PMID: 38532527 DOI: 10.1111/bjh.19422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/07/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Vaccinations are fundamental tools in preventing infectious diseases, especially in immunocompromised patients like those affected by non-Hodgkin lymphomas (NHLs). The COVID-19 pandemic made clinicians increasingly aware of the importance of vaccinations in preventing potential life-threatening SARS-CoV-2-related complications in NHL patients. However, several studies have confirmed a significant reduction in vaccine-induced immune responses after anti-CD20 monoclonal antibody treatment, thus underscoring the need for refined immunization strategies in NHL patients. In this review, we summarize the existing data about COVID-19 and other vaccine's efficacy in patients with NHL and propose multidisciplinary team-based recommendations for the management of vaccines in this specific group of patients.
Collapse
Affiliation(s)
- Michele Merli
- Division of Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Costantini
- Clinical Immunology Unit, Azienda Ospedaliero Universitaria delle Marche - Università Politecnica delle Marche, Ancona, Italy
| | - Silvio Tafuri
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Davide Fiore Bavaro
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, Aldo Moro University of Bari, Bari, Italy
| | - Carla Minoia
- Hematology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Erika Meli
- Division of Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Stefano Luminari
- Hematology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Surgical Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Guido Gini
- Clinic of Hematology, Azienda Ospedaliero Universitaria Delle Marche - Università Politecnica Delle Marche, Ancona, Italy
| |
Collapse
|
8
|
Moreno-Echevarria FM, Caputo MT, Camp DM, Reddy S, Achenbach CJ. Incidence and risk factors of omicron variant SARS-CoV-2 breakthrough infection among vaccinated and boosted individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.03.24305293. [PMID: 38633797 PMCID: PMC11023664 DOI: 10.1101/2024.04.03.24305293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Background SARS-CoV-2 vaccines have been shown to be safe and effective against infection and severe COVID-19 disease worldwide. Certain co-morbid conditions cause immune dysfunction and may reduce immune response to vaccination. In contrast, those with co-morbidities may practice infection prevention strategies. Thus, the real-world clinical impact of co-morbidities on SARS-CoV-2 infection in the recent post-vaccination period is not well established. We performed this study to understand the epidemiology of Omicron breakthrough infection and evaluate associations with number of comorbidities in a vaccinated and boosted population. Methods and Findings We performed a retrospective clinical cohort study utilizing the Northwestern Medicine Enterprise Data Warehouse. Our study population was identified as fully vaccinated adults with at least one booster. The primary risk factor of interest was the number of co-morbidities. Our primary outcome was incidence and time to first positive SARS-CoV-2 molecular test in the Omicron predominant era. We performed multivariable analyses stratified by calendar time using Cox modeling to determine hazard of SARS-CoV-2. In total, 133,191 patients were analyzed. Having 3+ comorbidities was associated with increased hazard for breakthrough (HR=1.2 CI 1.2-1.6). During the second half of the study, having 2 comorbidities (HR= 1.1 95% CI 1.02-1.2) and having 3+ comorbidities (HR 1.7, 95% CI 1.5-1.9) were associated with increased hazard for Omicron breakthrough. Older age was associated with decreased hazard in the first 6 months of follow-up. Interaction terms for calendar time indicated significant changes in hazard for many factors between the first and second halves of the follow-up period. Conclusions Omicron breakthrough is common with significantly higher risk for our most vulnerable patients with multiple co-morbidities. Age related behavioral factors play an important role in breakthrough infection with the highest incidence among young adults. Our findings reflect real-world differences in immunity and exposure risk behaviors for populations vulnerable to COVID-19.
Collapse
Affiliation(s)
| | - Mathew T. Caputo
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University
| | - Daniel M. Camp
- Robert J. Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University
| | - Susheel Reddy
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University
| | - Chad J. Achenbach
- Robert J. Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University
- Department of Medicine, Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University
| |
Collapse
|
9
|
El-Ashwah S, Salmanton-García J, Bilgin YM, Itri F, Žák P, Weinbergerová B, Verga L, Omrani AS, Silva MGD, Szotkowski T, Marchetti M, Buquicchio C, Nucci M, Schönlein M, Farina F, Besson C, Prezioso L, Nizamuddin S, Dávila-Valls J, Martín-Pérez S, Bonuomo V, Van Doesum J, Tisi MC, Passamonti F, Méndez GA, Meers S, Maertens J, López-García A, Glenthøj A, Bonnani M, Rinaldi I, Ormazabal-Vélez I, Labrador J, Kulasekararaj A, Espigado I, Demirkan F, De Jonge N, Collins GP, Calbacho M, Blennow O, Al-Khabori M, Adžić-Vukičević T, Arellano E, Mišković B, Mladenović M, Nordlander A, Ráčil Z, Ammatuna E, Cordoba R, Hersby DS, Gräfe S, Emarah Z, Hanakova M, Sacchi MV, Ijaz M, Rahimli L, Nunes Rodrigues R, Zambrotta GPM, Marchesi F, Cornely OA, Pagano L. The mortality of COVID-19 in CML patients from 2020 until 2022: results from the EPICOVIDEHA survey. Leuk Lymphoma 2024; 65:199-208. [PMID: 37966980 DOI: 10.1080/10428194.2023.2280886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/10/2023] [Indexed: 11/17/2023]
Abstract
Since the beginning of the COVID-19 pandemic, there has been an overall improvement in patient mortality. However, haematological malignancy patients continue to experience significant impacts from COVID-19, including high rates of hospitalization, intensive care unit (ICU) admissions, and mortality. In comparison to other haematological malignancy patients, individuals with chronic myeloid leukemia (CML) generally have better prognosis. This study, conducted using a large haematological malignancy patient database (EPICOVIDEHA), demonstrated that the majority of CML patients experienced mild infections. The decline in severe and critical infections over the years can largely be attributed to the widespread administration of vaccinations and the positive response they elicited. Notably, the mortality rate among CML patients was low and exhibited a downward trend in subsequent years. Importantly, our analysis provided confirmation of the effectiveness of vaccinations in CML patients.
Collapse
Affiliation(s)
| | - Jon Salmanton-García
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD) and Excellence Center for Medical Mycology (ECMM), University of Cologne, University Hospital Cologne, Cologne, Germany
| | - Yavuz M Bilgin
- Department of Internal Medicine, ADRZ, Goes, Netherlands
| | - Federico Itri
- San Luigi Gonzaga Hospital - Orbassano, Orbassano, Italy
| | - Pavel Žák
- University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Barbora Weinbergerová
- Department of Internal Medicine - Hematology and Oncology, Masaryk University Hospital Brno, Brno, Czech Republic
| | - Luisa Verga
- Azienda Ospedaliera San Gerardo - Monza, Monza, Italy
- Università Milano-Bicocca, Milan, Italy
| | - Ali S Omrani
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | | | | | - Monia Marchetti
- Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | | | - Marcio Nucci
- Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martin Schönlein
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Caroline Besson
- Centre Hospitalier de Versailles, Le Chesnay, France
- Université Paris-Saclay, UVSQ, Inserm, Équipe "Exposome et Hérédité", CESP, Villejuif, France
| | - Lucia Prezioso
- Hospital University of Parma - Hematology and Bone Marrow Unit, Parma, Italy
| | | | | | | | - Valentina Bonuomo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | | | - Francesco Passamonti
- Department of Medicine and Surgery, University of Insubria and ASST Sette Laghi, Ospedale di Circolo of Varese, Varese, Italy
| | | | | | - Johan Maertens
- Department of Microbiology, Immunology, and Transplantation, KULeuven, Leuven, Belgium
- Department of Hematology, UZ Leuven, Leuven, Belgium
| | - Alberto López-García
- Fundacion Jimenez Diaz University Hospital, Health Research Institute IIS-FJD, Madrid, Spain
| | - Andreas Glenthøj
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Matteo Bonnani
- Hematology Unit, Fondazione Policlinico Universitario Agostino Gemelli - IRCCS, Rome, Italy
| | - Ikhwan Rinaldi
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | | | - Jorge Labrador
- Department of Hematology, Research Unit, Hospital Universitario de Burgos, Burgos, Spain
| | | | - Ildefonso Espigado
- Department of Hematology, University Hospital Virgen Macarena - University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla (Departamento de Medicina), Seville, Spain
| | - Fatih Demirkan
- Division of Hematology, Dokuz Eylul University, Izmir, Turkey
| | - Nick De Jonge
- Amsterdam UMC, Location VUmc, Amsterdam, Netherlands
| | - Graham P Collins
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | | | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Elena Arellano
- Department of Hematology, University Hospital Virgen Macarena - University Hospital Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC), Universidad de Sevilla (Departamento de Medicina), Seville, Spain
| | - Bojana Mišković
- COVID-19 Hospital "Batajnica", Belgrade, Serbia
- Clinic for Orthopedic Surgery and Traumatology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Miloš Mladenović
- COVID-19 Hospital "Batajnica", Belgrade, Serbia
- Clinic for Orthopedic Surgery and Traumatology, University Clinical Center of Serbia, Belgrade, Serbia
| | - Anna Nordlander
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Zdeněk Ráčil
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | - Raul Cordoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute IIS-FJD, Madrid, Spain
| | - Ditte Stampe Hersby
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Stefanie Gräfe
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ziad Emarah
- Oncology Center, Mansoura University, Mansoura, Egypt
| | - Michaela Hanakova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Maria Vittoria Sacchi
- Azienda Ospedaliera Nazionale SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Marriyam Ijaz
- Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, Pakistan
| | - Laman Rahimli
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
| | | | | | - Francesco Marchesi
- Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Oliver A Cornely
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, University Hospital Cologne, Institute of Translational Research, Cologne, Germany
- Clinical Trials Centre Cologne (ZKS Köln), University of Cologne, University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine, and University Hospital Cologne, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Livio Pagano
- Hematology Unit, Fondazione Policlinico Universitario Agostino Gemelli - IRCCS, Rome, Italy
- Hematology Unit, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
10
|
Fowler T, Fellows A, MacISaac R, Kolade O, Singh B, Eccles A, Blandford E, Tunkel SA. The evaluation of a multi-day testing approach with antigen lateral flow devices for people eligible for community-based COVID-19 treatments. J Antimicrob Chemother 2023; 78:ii12-ii17. [PMID: 37995356 PMCID: PMC10666983 DOI: 10.1093/jac/dkad313] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND COVID-19 therapeutics including antiviral and monoclonal antibody treatments (hereafter 'COVID-19 treatments') require rapid administration to be effective. As part of the community-based antiviral and therapeutic treatment pathway for COVID-19 there has been a move from PCR testing in those eligible to a rapid antigen lateral flow testing regime. OBJECTIVES To determine whether a multi-day lateral flow device (LFD) testing regime is a feasible alternative to PCR for diagnosing symptomatic patients eligible for COVID-19 treatments. An LFD regime might return a positive result more quickly than a PCR and hence expedite access to COVID-19 treatments. METHODS A retrospective analysis was conducted of diagnostic testing for SARS-CoV-2 with a combination of PCR and LFDs of symptomatic patients eligible for COVID-19 treatments. LFD testing patterns were not assigned. Patients self-censored and the patterns were retro-fitted to the observed results. RESULTS The LFD testing patterns offered high sensitivity, close to 92%; however, the false positive rate also increased, with most of the multi-day testing patterns having a false positive rate greater than 3%. The highest sensitivity was seen among patients who tested with LFD on the same day as PCR. CONCLUSIONS There were multiple observed testing behaviours. We conclude that multi-day LFD testing for COVID-19 provides a feasible alternative to PCR to in eligible patients, allowing swift prescription of COVID-19 treatments in most cases. This approach requires acceptance of a trade-off for a small increase in false-positive and -negative results.
Collapse
Affiliation(s)
- Tom Fowler
- Clinical and Public Health Group, UK Health Security Agency, Nobel House, 17 Smith Square, London, SW1P 3JR, UK
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | | | - Olumide Kolade
- Clinical and Public Health Group, UK Health Security Agency, Nobel House, 17 Smith Square, London, SW1P 3JR, UK
| | - Bhupinder Singh
- Clinical and Public Health Group, UK Health Security Agency, Nobel House, 17 Smith Square, London, SW1P 3JR, UK
| | - Adam Eccles
- Clinical and Public Health Group, UK Health Security Agency, Nobel House, 17 Smith Square, London, SW1P 3JR, UK
| | - Edward Blandford
- Clinical and Public Health Group, UK Health Security Agency, Nobel House, 17 Smith Square, London, SW1P 3JR, UK
| | - Sarah A Tunkel
- Clinical and Public Health Group, UK Health Security Agency, Nobel House, 17 Smith Square, London, SW1P 3JR, UK
| |
Collapse
|
11
|
Kampouri E, Hill JA, Dioverti V. COVID-19 after hematopoietic cell transplantation and chimeric antigen receptor (CAR)-T-cell therapy. Transpl Infect Dis 2023; 25 Suppl 1:e14144. [PMID: 37767643 DOI: 10.1111/tid.14144] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
More than 3 years have passed since Coronavirus disease 2019 (COVID-19) was declared a global pandemic, yet COVID-19 still severely impacts immunocompromised individuals including those treated with hematopoietic cell transplantation (HCT) and chimeric antigen receptor-T-cell therapies who remain at high risk for severe COVID-19 and mortality. Despite vaccination efforts, these patients have inadequate responses due to immunosuppression, which underscores the need for additional preventive approaches. The optimal timing, schedule of vaccination, and immunological correlates for protective immunity remain unknown. Antiviral therapies used early during disease can reduce mortality and severity due to COVID-19. The combination or sequential use of antivirals could be beneficial to control replication and prevent the development of treatment-related mutations in protracted COVID-19. Despite conflicting data, COVID-19 convalescent plasma remains an option in immunocompromised patients with mild-to-moderate disease to prevent progression. Protracted COVID-19 has been increasingly recognized among these patients and has been implicated in intra-host emergence of SARS-CoV-2 variants. Finally, novel SARS-CoV2-specific T-cells and natural killer cell-boosting (or -containing) products may be active against multiple variants and are promising therapies in immunocompromised patients.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Veronica Dioverti
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Franceschini E, Pellegrino M, Todisco V, Dolci G, Bettelli F, Meschiari M, Bedini A, Fregni-Serpini G, Grottola A, Guaraldi G, Pecorari M, Sarti M, Luppi M, Perno CF, Mussini C. Persistent SARS-CoV-2 infection with multiple clinical relapses in two patients with follicular lymphoma treated with bendamustine and obinutuzumab or rituximab. Infection 2023; 51:1577-1581. [PMID: 37076752 PMCID: PMC10115373 DOI: 10.1007/s15010-023-02039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
PURPOSE People with hematologic malignancies have a significantly higher risk of developing severe and protracted forms of SARS-CoV-2 infection compared to immunocompetent patients, regardless of vaccination status. RESULTS We describe two cases of prolonged SARS-CoV-2 infection with multiple relapses of COVID-19 pneumonia in patients with follicular lymphoma treated with bendamustine and obinutuzumab or rituximab. The aim is to highlight the complexity of SARS-CoV-2 infection in this fragile group of patients and the necessity of evidence-based strategies to treat them properly. CONCLUSIONS Patients with hematological malignancies treated with bendamustine and anti-CD20 antibodies had a significant risk of prolonged and relapsing course of COVID-19. Specific preventive and therapeutic strategies should be developed for this group of patients.
Collapse
Affiliation(s)
- Erica Franceschini
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy.
| | - Mariachiara Pellegrino
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy
| | - Vera Todisco
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy
| | - Giovanni Dolci
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy
| | - Francesca Bettelli
- Hematology Unit and Chair, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - Marianna Meschiari
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy
| | - Andrea Bedini
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy
| | - Giulia Fregni-Serpini
- Molecular Microbiology and Virology Unit, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - Antonella Grottola
- Molecular Microbiology and Virology Unit, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy
| | - Monica Pecorari
- Molecular Microbiology and Virology Unit, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - Mario Sarti
- Clinical Microbiology Unit, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - Mario Luppi
- Hematology Unit and Chair, Azienda Ospedaliera Universitaria di Modena, Modena, Italy
| | - Carlo Federico Perno
- Unit of Diagnostic Microbiology and Immunology and Multimodal Medicine Area, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cristina Mussini
- Infectious Disease Unit, Azienda Ospedaliera Universitaria di Modena, Largo del Pozzo 71, 41124, Modena, Italy
| |
Collapse
|
13
|
Meejun T, Srisurapanont K, Manothummetha K, Thongkam A, Mejun N, Chuleerarux N, Sanguankeo A, Phongkhun K, Leksuwankun S, Thanakitcharu J, Lerttiendamrong B, Langsiri N, Torvorapanit P, Worasilchai N, Plongla R, Hirankarn N, Nematollahi S, Permpalung N, Moonla C, Kates OS. Attenuated immunogenicity of SARS-CoV-2 vaccines and risk factors in stem cell transplant recipients: a meta-analysis. Blood Adv 2023; 7:5624-5636. [PMID: 37389818 PMCID: PMC10514108 DOI: 10.1182/bloodadvances.2023010349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/05/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023] Open
Abstract
Immunogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is diminished in hematopoietic stem cell transplant (HSCT) recipients. To summarize current evidence and identify risk factors for attenuated responses, 5 electronic databases were searched since database inceptions through 12 January 2023 for studies reporting humoral and/or cellular immunogenicity of SARS-CoV-2 vaccination in the HSCT population. Using descriptive statistics and random-effects models, extracted numbers of responders and pooled odds ratios (pORs) with 95% confidence intervals (CIs) for risk factors of negative immune responses were analyzed (PROSPERO: CRD42021277109). From 61 studies with 5906 HSCT recipients, after 1, 2, and 3 doses of messenger RNA (mRNA) SARS-CoV-2 vaccines, the mean antispike antibody seropositivity rates (95% CI) were 38% (19-62), 81% (77-84), and 80% (75-84); neutralizing antibody seropositivity rates were 52% (40-64), 71% (54-83), and 78% (61-89); and cellular immune response rates were 52% (39-64), 66% (51-79), and 72% (52-86). After 2 vaccine doses, risk factors (pOR; 95% CI) associated with antispike seronegativity were male recipients (0.63; 0.49-0.83), recent rituximab exposure (0.09; 0.03-0.21), haploidentical allografts (0.46; 0.22-0.95), <24 months from HSCT (0.25; 0.07-0.89), lymphopenia (0.18; 0.13-0.24), hypogammaglobulinemia (0.23; 0.10-0.55), concomitant chemotherapy (0.48; 0.29-0.78) and immunosuppression (0.18; 0.13-0.25). Complete remission of underlying hematologic malignancy (2.55; 1.05-6.17) and myeloablative conditioning (1.72; 1.30-2.28) compared with reduced-intensity conditioning were associated with antispike seropositivity. Ongoing immunosuppression (0.31; 0.10-0.99) was associated with poor cellular immunogenicity. In conclusion, attenuated humoral and cellular immune responses to mRNA SARS-CoV-2 vaccination are associated with several risk factors among HSCT recipients. Optimizing individualized vaccination and developing alternative COVID-19 prevention strategies are warranted.
Collapse
Affiliation(s)
- Tanaporn Meejun
- Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Kasama Manothummetha
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Achitpol Thongkam
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nuthchaya Mejun
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nipat Chuleerarux
- Department of Medicine, University of Miami/Jackson Memorial Hospital, Miami, FL
| | - Anawin Sanguankeo
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kasidis Phongkhun
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Surachai Leksuwankun
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | | | - Nattapong Langsiri
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pattama Torvorapanit
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | - Rongpong Plongla
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Saman Nematollahi
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ
| | - Nitipong Permpalung
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chatphatai Moonla
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Olivia S. Kates
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
14
|
Nakahara H, Cheedarla N, Verkerke HP, Cheedarla S, Wu SC, Hendrickson JE, Chang A, McLemore ML, El Rassi F, Roback JD, Neish AS, Fasano RM, Stowell SR. Enhanced IgG immune response to COVID-19 vaccination in patients with sickle cell disease. Br J Haematol 2023; 202:937-941. [PMID: 37287128 PMCID: PMC10751105 DOI: 10.1111/bjh.18899] [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: 03/28/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023]
Abstract
Patients with sickle cell disease (SCD) are considered to be immunocompromised, yet data on the antibody response to SARS-CoV-2 vaccination in SCD is limited. We investigated anti-SARS-CoV-2 IgG titres and overall neutralizing activity in 201 adults with SCD and demographically matched non-SCD controls. Unexpectedly, patients with SCD generate a more robust and durable COVID-19 vaccine IgG response compared to matched controls, though the neutralizing activity remained similar across both cohorts. These findings suggest that patients with SCD achieve a similar antibody response following COVID-19 vaccination compared to the general population, with implications for optimal vaccination strategies for patients with SCD.
Collapse
Affiliation(s)
- Hirotomo Nakahara
- Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Narayanaiah Cheedarla
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hans P Verkerke
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Suneethamma Cheedarla
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shang-Chuen Wu
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeanne E Hendrickson
- Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Andres Chang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Morgan L McLemore
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Fuad El Rassi
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - John D Roback
- Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ross M Fasano
- Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Huang CT, Lee CP, Chen TY, Liu YC, Cho SF, Du JS, Yu ML, Huang CF, Wang SF, Hsiao HH. Serological Responses and Predictive Factors of Booster COVID-19 Vaccines in Patients with Hematologic Malignancies. J Clin Med 2023; 12:5647. [PMID: 37685720 PMCID: PMC10488979 DOI: 10.3390/jcm12175647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/01/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Patients with hematologic malignancies are reported to have a more severe course of coronavirus disease 2019 (COVID-19) and be less responsive to vaccination. In this prospective study, we aimed to evaluate the serological responses to booster COVID-19 vaccines of Taiwanese patients with hematologic malignancies and identify potential predictive markers for effective neutralizing immunity. This study enrolled 68 patients with hematologic malignancies and 68 age- and gender-matched healthy control subjects who received three doses of vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from 1 January 2022 to 31 October 2022. The SARS-CoV-2 immunoglobulin G (IgG) spike antibody level was measured with the Abbott assay. The effective neutralization capacity was defined as an anti-spike IgG level of ≥4160 AU/mL. Among the 68 patients with hematologic malignancies, 89.7% achieved seroconversion after booster doses. Seven patients with actively treated lymphoma remained seronegative and had the lowest humoral responses among patients with different types of hematologic malignancies. Despite comparable antibody titers between patients and healthy individuals, rates of effective neutralization (66.2% vs. 86.8%, respectively; p = 0.005) were significantly reduced in patients with hematologic malignancies. In a multivariate analysis, the independent predictors for effective neutralization were a lack of B-cell-targeted agents within six months of vaccination (odds ratio, 15.2; 95% confidence interval, 2.7-84.2; p = 0.002) and higher immunoglobulin levels (odds ratio, 4.4; 95% confidence interval, 1.3-14.7; p = 0.017). In conclusion, the majority of patients with hematologic malignancies achieved seroconversion after booster vaccination. Patients with ongoing B-cell depletion and hypogammaglobinemia were identified as having negative predictive markers for effective neutralization.
Collapse
Affiliation(s)
- Chien-Tzu Huang
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.H.); (C.-P.L.); (Y.-C.L.); (S.-F.C.); (J.-S.D.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ching-Ping Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.H.); (C.-P.L.); (Y.-C.L.); (S.-F.C.); (J.-S.D.)
| | - Tzu-Yin Chen
- Department of Nursing, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yi-Chang Liu
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.H.); (C.-P.L.); (Y.-C.L.); (S.-F.C.); (J.-S.D.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Feng Cho
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.H.); (C.-P.L.); (Y.-C.L.); (S.-F.C.); (J.-S.D.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jeng-Shiun Du
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.H.); (C.-P.L.); (Y.-C.L.); (S.-F.C.); (J.-S.D.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Lung Yu
- Division of Hepatobiliary, Department of Internal Medicine and Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.Y.); (C.-F.H.)
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Chung-Feng Huang
- Division of Hepatobiliary, Department of Internal Medicine and Hepatitis Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.Y.); (C.-F.H.)
- Ph.D. Program in Translational Medicine, College of Medicine, Kaohsiung Medical University and Academia Sinica, Kaohsiung 807, Taiwan
| | - Sheng-Fan Wang
- Center for Tropical Medicine and Infectious Disease Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hui-Hua Hsiao
- Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.H.); (C.-P.L.); (Y.-C.L.); (S.-F.C.); (J.-S.D.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
16
|
Chen P, Bergman P, Blennow O, Hansson L, Mielke S, Nowak P, Söderdahl G, Österborg A, Smith CIE, Vesterbacka J, Wullimann D, Cuapio A, Akber M, Bogdanovic G, Muschiol S, Åberg M, Loré K, Sällberg Chen M, Buggert M, Ljungman P, Aleman S, Ljunggren HG. Real-world assessment of immunogenicity in immunocompromised individuals following SARS-CoV-2 mRNA vaccination: a one-year follow-up of the prospective clinical trial COVAXID. EBioMedicine 2023; 94:104700. [PMID: 37453361 PMCID: PMC10365982 DOI: 10.1016/j.ebiom.2023.104700] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Immunocompromised patients have varying responses to SARS-CoV-2 mRNA vaccination. However, there is limited information available from prospective clinical trial cohorts with respect to long-term immunogenicity-related responses in these patient groups following three or four vaccine doses, and in applicable cases infection. METHODS In a real-world setting, we assessed the long-term immunogenicity-related responses in patients with primary and secondary immunodeficiencies from the prospective open-label clinical trial COVAXID. The original clinical trial protocol included two vaccine doses given on days 0 and 21, with antibody titres measured at six different timepoints over six months. The study cohort has subsequently been followed for one year with antibody responses evaluated in relation to the third and fourth vaccine dose, and in applicable cases SARS-CoV-2 infection. In total 356/539 patients were included in the extended cohort. Blood samples were analysed for binding antibody titres and neutralisation against the Spike protein for all SARS-CoV-2 variants prevailing during the study period, including Omicron subvariants. SARS-CoV-2 infections that did not require hospital care were recorded through quarterly in-person, or phone-, interviews and assessment of IgG antibody titres against SARS-CoV-2 Nucleocapsid. The original clinical trial was registered in EudraCT (2021-000175-37) and clinicaltrials.gov (NCT04780659). FINDINGS The third vaccine dose significantly increased Spike IgG titres against all the SARS-CoV-2 variants analysed in all immunocompromised patient groups. Similarly, neutralisation also increased against all variants studied, except for Omicron. Omicron-specific neutralisation, however, increased after a fourth dose as well as after three doses and infection in many of the patient subgroups. Noteworthy, however, while many patient groups mounted strong serological responses after three and four vaccine doses, comparably weak responders were found among patient subgroups with specific primary immunodeficiencies and subgroups with immunosuppressive medication. INTERPRETATION The study identifies particularly affected patient groups in terms of development of long-term immunity among a larger group of immunocompromised patients. In particular, the results highlight poor vaccine-elicited neutralising responses towards Omicron subvariants in specific subgroups. The results provide additional knowledge of relevance for future vaccination strategies. FUNDING The present studies were supported by grants from the Swedish Research Council, the Knut and Alice Wallenberg Foundation, Nordstjernan AB, Region Stockholm, and Karolinska Institutet.
Collapse
Affiliation(s)
- Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine, Clinical Immunology, Karolinska Institutet, Stockholm, Sweden
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Lotta Hansson
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Mielke
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Karolinska Comprehensive Cancer Center, Stockholm, Sweden; Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Söderdahl
- Department of Transplantation, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - C I Edvard Smith
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jan Vesterbacka
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - David Wullimann
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Angelica Cuapio
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mira Akber
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gordana Bogdanovic
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Muschiol
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Loré
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Karolinska Comprehensive Cancer Center, Stockholm, Sweden; Division of Hematology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
17
|
Ketkar A, Willey V, Pollack M, Glasser L, Dobie C, Wenziger C, Teng CC, Dube C, Cunningham D, Verduzco-Gutierrez M. Assessing the risk and costs of COVID-19 in immunocompromised populations in a large United States commercial insurance health plan: the EPOCH-US Study. Curr Med Res Opin 2023; 39:1103-1118. [PMID: 37431293 DOI: 10.1080/03007995.2023.2233819] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/12/2023]
Abstract
OBJECTIVE To estimate the prevalence of patients with an immunocompromising condition at risk for COVID-19, estimate COVID-19 prevalence rate (PR) and incidence rate (IR) by immunocompromising condition, and describe COVID-19-related healthcare resource utilization (HCRU) and costs. METHODS Using the Healthcare Integrated Research Database (HIRD), patients with ≥1 claim for an immunocompromising condition of interest or ≥2 claims for an immunosuppressive (IS) treatment and COVID-19 diagnosis during the infection period (1 April 2020-31 March 2022) and had ≥12 months baseline data were included. Cohorts (other than the composite cohort) were not mutually exclusive and were defined by each immunocompromising condition. Analyses were descriptive in nature. RESULTS Of the 16,873,161 patients in the source population, 2.7% (n = 458,049) were immunocompromised (IC). The COVID-19 IR for the composite IC cohort during the study period was 101.3 per 1000 person-years and the PR was 13.5%. The highest IR (195.0 per 1000 person-years) and PR (20.1%) were seen in the end-stage renal disease (ESRD) cohort; the lowest IR (68.3 per 1000 person-years) and PR (9.4%) were seen in the hematologic or solid tumor malignancy cohort. Mean costs for hospitalizations associated with the first COVID-19 diagnosis were estimated at nearly $1 billion (2021 United States dollars [USD]) for 14,516 IC patients, with a mean cost of $64,029 per patient. CONCLUSIONS Immunocompromised populations appear to be at substantial risk of severe COVID-19 outcomes, leading to increased costs and HCRU. Effective prophylactic options are still needed for these high-risk populations as the COVID-19 landscape evolves.
Collapse
Affiliation(s)
| | | | | | - Lisa Glasser
- AstraZeneca Biopharmaceuticals Medical, Wilmington, DE, USA
| | | | | | - Chia-Chen Teng
- AstraZeneca Biopharmaceuticals Medical, Wilmington, DE, USA
| | - Christine Dube
- AstraZeneca Biopharmaceuticals Medical, Wilmington, DE, USA
| | | | | |
Collapse
|
18
|
Hofsink Q, Haggenburg S, Lissenberg-Witte BI, Broers AEC, van Doesum JA, van Binnendijk RS, den Hartog G, Bhoekhan MS, Haverkate NJE, van Meerloo J, Burger JA, Bouhuijs JH, Smits GP, Wouters D, van Leeuwen EMM, Bontkes HJ, Kootstra NA, Vogels-Nooijen S, Rots N, van Beek J, Heemskerk MHM, Groen K, van Meerten T, Mutsaers PGNJ, van Gils MJ, Goorhuis A, Rutten CE, Hazenberg MD, Nijhof IS. Fourth mRNA COVID-19 vaccination in immunocompromised patients with haematological malignancies (COBRA KAI): a cohort study. EClinicalMedicine 2023; 61:102040. [PMID: 37337616 PMCID: PMC10270678 DOI: 10.1016/j.eclinm.2023.102040] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/21/2023] Open
Abstract
Background Patients with haematological malignancies have impaired antibody responses to SARS-CoV-2 vaccination. We aimed to investigate whether a fourth mRNA COVID-19 vaccination improved antibody quantity and quality. Methods In this cohort study, conducted at 5 sites in the Netherlands, we compared antibody concentrations 28 days after 4 mRNA vaccinations (3-dose primary series plus 1 booster vaccination) in SARS-CoV-2 naive, immunocompromised patients with haematological malignancies to those obtained by age-matched, healthy individuals who had received the standard primary 2-dose mRNA vaccination schedule followed by a first booster mRNA vaccination. Prior to and 4 weeks after each vaccination, peripheral blood samples and data on demographic parameters and medical history were collected. Concentrations of antibodies that bind spike 1 (S1) and nucleocapsid (N) protein of SARS-CoV-2 were quantified in binding antibody units (BAU) per mL according to the WHO International Standard for COVID-19 serological tests. Seroconversion was defined as an S1 IgG concentration >10 BAU/mL and a previous SARS-CoV-2 infection as N IgG >14.3 BAU/mL. Antibody neutralising activity was tested using lentiviral-based pseudoviruses expressing spike protein of SARS-CoV-2 wild-type (D614G), Omicron BA.1, and Omicron BA.4/5 variants. This study is registered with EudraCT, number 2021-001072-41. Findings Between March 24, 2021 and May 4, 2021, 723 patients with haematological diseases were enrolled, of which 414 fulfilled the inclusion criteria for the current analysis. Although S1 IgG concentrations in patients significantly improved after the fourth dose, they remained significantly lower compared to those obtained by 58 age-matched healthy individuals after their first booster (third) vaccination. The rise in neutralising antibody concentration was most prominent in patients with a recovering B cell compartment, although potent responses were also observed in patients with persistent immunodeficiencies. 19% of patients never seroconverted, despite 4 vaccinations. Patients who received their first 2 vaccinations when they were B cell depleted and the third and fourth vaccination during B cell recovery demonstrated similar antibody induction dynamics as patients with normal B cell numbers during the first 2 vaccinations. However, the neutralising capacity of these antibodies was significantly better than that of patients with normal B cell numbers after two vaccinations. Interpretation A fourth mRNA COVID-19 vaccination improved S1 IgG concentrations in the majority of patients with a haematological malignancy. Vaccination during B cell depletion may pave the way for better quality of antibody responses after B cell reconstitution. Funding The Netherlands Organisation for Health Research and Development and Amsterdam UMC.
Collapse
Affiliation(s)
- Quincy Hofsink
- Department of Haematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
| | - Sabine Haggenburg
- Department of Haematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
| | - Birgit I Lissenberg-Witte
- Department of Epidemiology and Data Science, Amsterdam UMC Location Vrije Universiteit, Amsterdam, Netherlands
| | - Annoek E C Broers
- Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Jaap A van Doesum
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Rob S van Binnendijk
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Gerco den Hartog
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, Netherlands
- Laboratory of Medical Immunology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Michel S Bhoekhan
- Department of Haematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
| | - Nienke J E Haverkate
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Johan van Meerloo
- Department of Haematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, Netherlands
- Cancer Centre Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Judith A Burger
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Joey H Bouhuijs
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Gaby P Smits
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Dorine Wouters
- Central Diagnostic Laboratory, Amsterdam UMC, Amsterdam, Netherlands
| | - Ester M M van Leeuwen
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Hetty J Bontkes
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Department of Clinical Chemistry, Laboratory Medical Immunology, Amsterdam UMC, Amsterdam, Netherlands
| | - Neeltje A Kootstra
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | | | - Nynke Rots
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Josine van Beek
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | | | - Kazimierz Groen
- Department of Haematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, Netherlands
| | - Tom van Meerten
- Department of Haematology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Pim G N J Mutsaers
- Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Marit J van Gils
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Abraham Goorhuis
- Department of Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Caroline E Rutten
- Department of Haematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Mette D Hazenberg
- Department of Haematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, Netherlands
- Cancer Centre Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
- Department of Haematopoiesis, Sanquin Research, Amsterdam, Netherlands
| | - Inger S Nijhof
- Department of Haematology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, Netherlands
- Department of Internal Medicine-Haematology, St. Antonius Hospital, Nieuwegein, Netherlands
| |
Collapse
|
19
|
Fattizzo B, Rampi N, Barcellini W. Vaccinations in hematological patients in the era of target therapies: Lesson learnt from SARS-CoV-2. Blood Rev 2023; 60:101077. [PMID: 37029066 PMCID: PMC10043962 DOI: 10.1016/j.blre.2023.101077] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/14/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023]
Abstract
Novel targeting agents for hematologic diseases often exert on- or off-target immunomodulatory effects, possibly impacting on response to anti-SARS-CoV-2 vaccinations and other vaccines. Agents that primarily affect B cells, particularly anti-CD20 monoclonal antibodies (MoAbs), Bruton tyrosine kinase inhibitors, and anti-CD19 chimeric antigen T-cells, have the strongest impact on seroconversion. JAK2, BCL-2 inhibitors and hypomethylating agents may hamper immunity but show a less prominent effect on humoral response to vaccines. Conversely, vaccine efficacy seems not impaired by anti-myeloma agents such as proteasome inhibitors and immunomodulatory agents, although lower seroconversion rates are observed with anti-CD38 and anti-BCMA MoAbs. Complement inhibitors for complement-mediated hematologic diseases and immunosuppressants used in aplastic anemia do not generally affect seroconversion rate, but the extent of the immune response is reduced under steroids or anti-thymocyte globulin. Vaccination is recommended prior to treatment or as far as possible from anti-CD20 MoAb (at least 6 months). No clearcut indications for interrupting continuous treatment emerged, and booster doses significantly improved seroconversion. Cellular immune response appeared preserved in several settings.
Collapse
Affiliation(s)
- Bruno Fattizzo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| | - Nicolò Rampi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Wilma Barcellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
20
|
Terpos E, Musto P, Engelhardt M, Delforge M, Cook G, Gay F, van de Donk NWCJ, Ntanasis-Stathopoulos I, Vangsted AJ, Driessen C, Schjesvold F, Cerchione C, Zweegman S, Hajek R, Moreau P, Einsele H, San-Miguel J, Boccadoro M, Dimopoulos MA, Sonneveld P, Ludwig H. Management of patients with multiple myeloma and COVID-19 in the post pandemic era: a consensus paper from the European Myeloma Network (EMN). Leukemia 2023:10.1038/s41375-023-01920-1. [PMID: 37142661 PMCID: PMC10157596 DOI: 10.1038/s41375-023-01920-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/06/2023]
Abstract
In the post-pandemic COVID-19 period, human activities have returned to normal and COVID-19 cases are usually mild. However, patients with multiple myeloma (MM) present an increased risk for breakthrough infections and severe COVID-19 outcomes, including hospitalization and death. The European Myeloma Network has provided an expert consensus to guide patient management in this era. Vaccination with variant-specific booster vaccines, such as the bivalent vaccine for the ancestral Wuhan strain and the Omicron BA.4/5 strains, is essential as novel strains emerge and become dominant in the community. Boosters should be administered every 6-12 months after the last vaccine shot or documented COVID-19 infection (hybrid immunity). Booster shots seem to overcome the negative effect of anti-CD38 monoclonal antibodies on humoral responses; however, anti-BCMA treatment remains an adverse predictive factor for humoral immune response. Evaluation of the immune response after vaccination may identify a particularly vulnerable subset of patients who may need additional boosters, prophylactic therapies and prevention measures. Pre-exposure prophylaxis with tixagevimab/cilgavimab is not effective against the new dominant variants and thus is no longer recommended. Oral antivirals (nirmatrelvir/ritonavir and molnupiravir) and remdesivir are effective against Omicron subvariants BA.2.12.1, BA.4, BA.5, BQ.1.1 and/or XBB.1.5 and should be administered in MM patients at the time of a positive COVID-19 test or within 5 days post symptoms onset. Convalescent plasma seems to have low value in the post-pandemic era. Prevention measures during SARS-CoV-2 outbreaks, including mask wearing and avoiding crowded places, seem prudent to continue for MM patients.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Pellegrino Musto
- Department of Precision and Regenerative Medicine and Ionian Area, "Aldo Moro" University School of Medicine, Bari, Italy
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy
| | - Monika Engelhardt
- Department of Hematology and Oncology, Interdisciplinary Cancer Center and Comprehensive Cancer Center Freiburg, University of Freiburg, Faculty of Freiburg, Freiburg, Germany
| | - Michel Delforge
- Department of Oncology, University Hospital Leuven, Leuven, Belgium
| | - Gordon Cook
- CRUK Clinical Trials Unit, Leeds Institute of Clinical Trial Research, University of Leeds, Leeds, UK
| | - Francesca Gay
- Division of Hematology, University of Turin, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Niels W C J van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Annette Juul Vangsted
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christoph Driessen
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Center for B-Cell Malignancies, University of Oslo, Oslo, Norway
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Roman Hajek
- Department of Hemato-Oncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Philippe Moreau
- Department of Hematology, University Hospital Hotel-Dieu, Nantes, France
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Jesus San-Miguel
- Cancer Center Clínica Universidad de Navarra, CCUN, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Mario Boccadoro
- Division of Hematology, University of Turin, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, First Department of Medicine, Center for Oncology, Hematology, and Palliative Care, Clinic Ottakring, Vienna, Austria
| |
Collapse
|
21
|
Schmidt KLJ, Dautzenberg NMM, Hoogerbrugge PM, Lindemans CA, Nierkens S, Smits G, Van Binnendijk RS, Bont LJ, Tissing WJE. Immune Response following BNT162b2 mRNA COVID-19 Vaccination in Pediatric Cancer Patients. Cancers (Basel) 2023; 15:cancers15092562. [PMID: 37174028 PMCID: PMC10177402 DOI: 10.3390/cancers15092562] [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: 03/23/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
COVID-19 vaccinations are recommended for children with cancer but data on their vaccination response is scarce. This study assesses the antibody and T-cell response following a 2- or 3-dose vaccination with BNT162b2 mRNA COVID-19 vaccine in children (5-17 years) with cancer. For the antibody response, participants with a serum concentration of anti-SARS-CoV-2 spike 1 antibodies of >300 binding antibody units per milliliter were classified as good responders. For the T-cell response, categorization was based on spike S1 specific interferon-gamma release with good responders having >200 milli-international units per milliliter. The patients were categorized as being treated with chemo/immunotherapy for less than 6 weeks (Tx < 6 weeks) or more than 6 weeks (Tx > 6 weeks) before the first immunization event. In 46 patients given a 2-dose vaccination series, the percentage of good antibody and good T-cell responders was 39.3% and 73.7% in patients with Tx < 6 weeks and 94.4% and 100% in patients with Tx > 6 weeks, respectively. An additional 3rd vaccination in 16 patients with Tx < 6 weeks, increased the percentage of good antibody responders to 70% with no change in T-cell response. A 3-dose vaccination series effectively boosted antibody levels and is of value for patients undergoing active cancer treatment.
Collapse
Affiliation(s)
- K L Juliëtte Schmidt
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Noël M M Dautzenberg
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Peter M Hoogerbrugge
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
| | - Caroline A Lindemans
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Gaby Smits
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Rob S Van Binnendijk
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Louis J Bont
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Wim J E Tissing
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands
- Department of Pediatric Oncology and Hematology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
22
|
Saavedra C, Vallejo A, Longo F, Serrano JJ, Fernández M, Gion M, López-Miranda E, Martínez-Jáñez N, Guerra E, Chamorro J, Rosero D, Velasco H, Martín A, Carrato A, Casado JL, Cortés A. Discordant Humoral and T-Cell Response to mRNA SARS-CoV-2 Vaccine and the Risk of Breakthrough Infections in Women with Breast Cancer, Receiving Cyclin-Dependent Kinase 4 and 6 Inhibitors. Cancers (Basel) 2023; 15:cancers15072000. [PMID: 37046661 PMCID: PMC10093435 DOI: 10.3390/cancers15072000] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/09/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Few data are available about the immune response to mRNA SARS-CoV-2 vaccines in patients with breast cancer receiving cyclin-dependent kinase 4/6 inhibitors (CDK4/6i). We conducted a prospective, single-center study of patients with breast cancer treated with CDK4/6i who received mRNA-1273 vaccination, as well as a comparative group of healthcare workers. The primary endpoint was to compare the rate and magnitude of humoral and T-cell response after full vaccination. A better neutralizing antibody and anti-S IgG level was observed after vaccination in the subgroup of women receiving CDK4/6i, but a trend toward a reduced CD4 and CD8 T-cell response in the CDK4/6i group was not statistically significant. There were no differences in the rate of COVID-19 after vaccination (19% vs. 12%), but breakthrough infections were observed in those with lower levels of anti-S IgG and neutralizing antibodies after the first dose. A lower rate of CD4 T-cell response was also found in those individuals with breakthrough infections, although a non-significant and similar level of CD8 T-cell response was also observed, regardless of breakthrough infections. The rate of adverse events was higher in patients treated with CDK4/6i, without serious adverse events. In conclusion, there was a robust humoral response, but a blunted T-cell response to mRNA vaccine in women receiving CDK4/6i, suggesting a reduced trend of the adaptative immune response.
Collapse
Affiliation(s)
- Cristina Saavedra
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Alejandro Vallejo
- Laboratory of Immunovirology, Department of Infectious Diseases, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Federico Longo
- CIBERONC, Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain
| | - Juan José Serrano
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - María Fernández
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - María Gion
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Elena López-Miranda
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Noelia Martínez-Jáñez
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Eva Guerra
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Jesús Chamorro
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Diana Rosero
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
| | - Héctor Velasco
- CiberInfect, Infectious Disease Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | - Adrián Martín
- CiberInfect, Infectious Disease Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
| | - Alfredo Carrato
- CIBERONC, Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain
| | - José Luis Casado
- CiberInfect, Infectious Disease Department, Ramón y Cajal University Hospital, 28034 Madrid, Spain
- Correspondence: (J.L.C.); (A.C.)
| | - Alfonso Cortés
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, 28034 Madrid, Spain
- Correspondence: (J.L.C.); (A.C.)
| |
Collapse
|
23
|
Ludwig H, Kumar S. Prevention of infections including vaccination strategies in multiple myeloma. Am J Hematol 2023; 98 Suppl 2:S46-S62. [PMID: 36251367 DOI: 10.1002/ajh.26766] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/12/2022]
Abstract
Infections are a major cause of morbidity and mortality in multiple myeloma. The increased risk for bacterial and viral infections results mainly from the disease-inherent and treatment-induced immunosuppression. Additional risk factors are older age with immune senescence, T cell depletion, polymorbidity, and male gender. Hence, every effort should be taken to reduce the risk for infections by identifying patients at higher risk for these complications and by implementing prophylactic measures, including chemoprophylaxis and immunization against various relevant pathogens. Here, we review the available evidence and provide recommendations for medical prophylaxis and vaccination in clinical practice.
Collapse
Affiliation(s)
- Heinz Ludwig
- Department of Medicine I, Center for Medical Oncology and Hematology with Outpatient Department and Palliative Care, Wilhelminen Cancer Research Institute, Vienna, Austria
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
24
|
Belsky JA, Carroll WR, Feliciano A, Jacob SA. Side effects following COVID-19 vaccination in pediatric patients with sickle cell disease. Pediatr Blood Cancer 2023; 70:e30193. [PMID: 36583456 PMCID: PMC9880642 DOI: 10.1002/pbc.30193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/31/2022]
Abstract
Vulnerable patient populations have seen decreased rates of vaccination against SARS-CoV2-19 (COVID-19) due to hesitancies and distrust, magnified by a paucity of data for certain populations. The rate of COVID-19 vaccination in children with sickle cell disease (SCD) remains low despite the risk for severe complications, resulting in continued infections and hospitalizations from COVID-19. We sought to describe vaccine reactions, including vaso-occlusive crises, emergency department visits, and hospitalizations, in children with SCD. Our findings will start to provide the necessary vaccine side effect data to inform patients, caregivers, and clinicians considering the COVID-19 primary vaccination series.
Collapse
Affiliation(s)
- Jennifer A. Belsky
- Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA,Division of Pediatric Hematology OncologyRiley Hospital for ChildrenIndianapolisIndianaUSA,Marian University College of Osteopathic MedicineIndianapolisIndianaUSA
| | | | - Anna Feliciano
- Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Seethal A. Jacob
- Department of PediatricsIndiana University School of MedicineIndianapolisIndianaUSA,Division of Pediatric Hematology OncologyRiley Hospital for ChildrenIndianapolisIndianaUSA,Center for Pediatric and Adolescent Comparative Effectiveness ResearchIndianapolisIndianaUSA
| |
Collapse
|
25
|
Luque-Paz D, Sesques P, Wallet F, Bachy E, Ader F. B-cell malignancies and COVID-19: a narrative review. Clin Microbiol Infect 2023; 29:332-337. [PMID: 36336236 PMCID: PMC9633106 DOI: 10.1016/j.cmi.2022.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND COVID-19 has been extensively characterized in immunocompetent hosts and to a lesser extent in immunocompromised populations. Among the latter, patients treated for B-cell malignancies have immunosuppression generated by B-cell lymphodepletion/aplasia resulting in an increased susceptibility to respiratory virus infections and poor response to vaccination. The consequence is that these patients are likely to develop severe or critical COVID-19. OBJECTIVES To examine the overall impact of COVID-19 in patients treated for a B-cell malignancy or receiving chimeric antigen receptor T (CAR-T) immunotherapy administered in case of relapsed or refractory disease. SOURCES We searched in the MEDLINE database to identify relevant studies, trials, reviews, or meta-analyses focusing on SARS-CoV-2 vaccination or COVID-19 management in patients treated for a B-cell malignancy or recipients of CAR-T cell therapy up to 8 July 2022. CONTENT The epidemiology and outcomes of COVID-19 in patients with B-cell malignancy and CAR-T cell recipients are summarized. Vaccine efficacy in these subgroups is compiled. Considering the successive surges of variants of concern, we propose a critical appraisal of treatment strategies by discussing the use of neutralizing monoclonal antibodies, convalescent plasma therapy, direct-acting antiviral drugs, corticosteroids, and immunomodulators. IMPLICATIONS For patients with B-cell malignancy, preventive vaccination against SARS-CoV-2 remains essential and the management of COVID-19 includes control of viral replication because of protracted SARS-CoV-2 shedding. Passive immunotherapy (monoclonal neutralizing antibody therapy and convalescent plasma therapy) and direct-active antivirals, such as remdesivir and nirmatrelvir/ritonavir are the best currently available treatments. Real-world data and subgroup analyses in larger trials are warranted to assess COVID-19 therapeutics in B-cell depleted populations.
Collapse
Affiliation(s)
- David Luque-Paz
- Université Rennes-I, Maladies Infectieuses et Réanimation Médicale, Hôpital Pontchaillou, Rennes, France
| | - Pierre Sesques
- Service d’Hématologie clinique, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Florent Wallet
- Service d'Anesthésie, médecine intensive, réanimation, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Emmanuel Bachy
- Service d’Hématologie clinique, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Florence Ader
- Département des Maladies infectieuses et tropicales, Hospices Civils de Lyon, F-69004, Lyon, France; Centre International de Recherche en Infectiologie (CIRI), Inserm 1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, F-69007, France.
| | | |
Collapse
|
26
|
Suzuki T, Kusumoto S, Kamezaki Y, Hashimoto H, Nishitarumizu N, Nakanishi Y, Kato Y, Kawai A, Matsunaga N, Ebina T, Nakamura T, Marumo Y, Oiwa K, Kinoshita S, Narita T, Ito A, Inagaki A, Ri M, Komatsu H, Aritsu T, Iida S. A comprehensive evaluation of humoral immune response to second and third SARS-CoV-2 mRNA vaccination in patients with malignant lymphoma. Int J Hematol 2023; 117:900-909. [PMID: 36790667 PMCID: PMC9930006 DOI: 10.1007/s12185-023-03550-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/16/2023]
Abstract
More information is needed regarding the efficacy of SARS-CoV-2 mRNA vaccines in immunocompromised populations, including patients with malignant lymphoma. This study aimed to evaluate humoral responses to the second and third mRNA vaccine doses in 165 lymphoma patients by retrospective analysis of serum SARS-CoV-2 spike protein antibody (S-IgG) titers. Patients with S-IgG titers ≥ 300, 10-300, and ≤ 10 binding antibody units (BAU)/mL were defined as adequate responders, low responders, and non-responders, respectively. S-IgG titers > 10 BAU/mL were considered to indicate seroconversion. After the second dose, 56%, 16%, and 28% of patients were adequate responders, low responders and non-responders, respectively. Multivariate analysis revealed that being an adequate responder after the second dose was associated with receiving the vaccine > 12 months after last chemotherapy, total peripheral lymphocyte count of ≥ 1000/µL, estimated glomerular filtration rate of ≥ 50 mL/min/1.73 m2, and vaccine type (mRNA-1273). After the third dose, patients had significantly higher S-IgG titers and a greater proportion achieved seroconversion. With this third dose, 26% of second-dose non-responders achieved seroconversion and 68% of second-dose low responders became adequate responders. Subsequent SARS-CoV-2 mRNA vaccinations may elicit an immune response in immunocompromised patients who do not initially respond to vaccination.
Collapse
Affiliation(s)
- Tomotaka Suzuki
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Shigeru Kusumoto
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi, Japan.
| | - Yoshiko Kamezaki
- grid.419812.70000 0004 1777 4627Scientific Information, Scientific Affairs, Sysmex Corporation, Kobe, Hyogo Japan
| | - Hiroya Hashimoto
- grid.411885.10000 0004 0469 6607Clinical Research Management Center, Nagoya City University Hospital, Nagoya, Japan
| | - Nozomi Nishitarumizu
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Yoko Nakanishi
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Yukiyasu Kato
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Akimi Kawai
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Naohiro Matsunaga
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Toru Ebina
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Tomoyuki Nakamura
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Yoshiaki Marumo
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Kana Oiwa
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Shiori Kinoshita
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Tomoko Narita
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Asahi Ito
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Atsushi Inagaki
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Masaki Ri
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Hirokazu Komatsu
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| | - Takashi Aritsu
- grid.419812.70000 0004 1777 4627Scientific Information, Scientific Affairs, Sysmex Corporation, Kobe, Hyogo Japan
| | - Shinsuke Iida
- grid.260433.00000 0001 0728 1069Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, Aichi Japan
| |
Collapse
|
27
|
Sejdic A, Frische A, Jørgensen CS, Rasmussen LD, Trebbien R, Dungu A, Holler JG, Ostrowski SR, Eriksson R, Søborg C, Nielsen TL, Fischer TK, Lindegaard B, Franck KT, Harboe ZB. High titers of neutralizing SARS-CoV-2 antibodies six months after symptom onset are associated with increased severity in COVID-19 hospitalized patients. Virol J 2023; 20:14. [PMID: 36698135 PMCID: PMC9875770 DOI: 10.1186/s12985-023-01974-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Viral shedding and neutralizing antibody (NAb) dynamics among patients hospitalized with severe coronavirus disease 2019 (COVID-19) and immune correlates of protection have been key questions throughout the pandemic. We investigated the duration of reverse transcriptase-polymerase chain reaction (RT-PCR) positivity, infectious viral shedding and NAb titers as well as the association between NAb titers and disease severity in hospitalized COVID-19 patients in Denmark 2020-2021. MATERIALS AND METHODS Prospective single-center observational cohort study of 47 hospitalized COVID-19 patients. Oropharyngeal swabs were collected at eight time points during the initial 30 days of inclusion. Serum samples were collected after a median time of 7 (IQR 5 - 10), 37 (IQR 35 - 38), 97 (IQR 95 - 100), and 187 (IQR 185 - 190) days after symptom onset. NAb titers were determined by an in-house live virus microneutralization assay. Viral culturing was performed in Vero E6 cells. RESULTS Patients with high disease severity had higher mean log2 NAb titers at day 37 (1.58, 95% CI [0.34 -2.81]), 97 (2.07, 95% CI [0.53-3.62]) and 187 (2.49, 95% CI [0.20- 4.78]) after symptom onset, compared to patients with low disease severity. Peak viral load (0.072, 95% CI [- 0.627 - 0.728]), expressed as log10 SARS-CoV-2 copies/ml, was not associated with disease severity. Virus cultivation attempts were unsuccessful in almost all (60/61) oropharyngeal samples collected shortly after hospital admission. CONCLUSIONS We document an association between high disease severity and high mean NAb titers at days 37, 97 and 187 after symptom onset. However, peak viral load during admission was not associated with disease severity. TRIAL REGISTRATION The study is registered at https://clinicaltrials.gov/ (NCT05274373).
Collapse
Affiliation(s)
- Adin Sejdic
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark.
- Statens Serum Institut, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | - Arnold Dungu
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Jon G Holler
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Robert Eriksson
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
- Department of Infectious Diseases, Karolinska Institutet, Solna, Sweden
| | - Christian Søborg
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Thyge L Nielsen
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Thea K Fischer
- Department of Clinical Research, Copenhagen University Hospital - North Zealand, Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Lindegaard
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Zitta Barrella Harboe
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
- Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
[Chinese consensus on severe acute respiratory syndrome coronavirus-2 vaccination in adult patients with hematological diseases (2023)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:19-25. [PMID: 36599437 PMCID: PMC10067370 DOI: 10.3760/cma.j.issn.0253-2727.2023.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Indexed: 01/06/2023]
|
29
|
Evaluation of Safety and Immunogenicity of a Recombinant Receptor-Binding Domain (RBD)-Tetanus Toxoid (TT) Conjugated SARS-CoV-2 Vaccine (PastoCovac) in Recipients of Autologous Hematopoietic Stem Cell Transplantation Compared to the Healthy Controls; A Prospective, Open-Label Clinical Trial. Vaccines (Basel) 2023; 11:vaccines11010117. [PMID: 36679963 PMCID: PMC9863563 DOI: 10.3390/vaccines11010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023] Open
Abstract
Background: The urgent need for prompt SARS-CoV-2 immunization of hematopoietic stem cell transplant (HSCT) recipients in an endemic area raises many challenges regarding selecting a vaccine platform appropriate for HSCT recipients being economical for widespread use in developing countries. Methods: The trial is a prospective, single-group, open-label study to investigate the safety and serologic response of two doses of the recombinant receptor-binding domain (RBD)-Tetanus Toxoid (TT) conjugated SARS-CoV-2 vaccine (PastoCovac) early after autologous (auto) HSCT. For this reason, a total of 38 patients who completed the two-dose SARS-CoV-2 RBD-based vaccine between three to nine months after auto-HSCT and had an available anti-spike serologic test at three predefined time points of baseline and after the first and second doses and 50 healthy control individuals were included in the analysis. The primary outcome was defined as an increase in IgG Immune status ratio (ISR) to the cut-off value for the positive result (≥1.1) in the semiquantitative test. Findings: The median time between auto-HSCT and vaccination was 127 days. No participant reported any significant adverse effects (Grade 3). Pain at the injection site was the most common adverse event. The ISR increased significantly (p < 0.001) during the three-time point sampling for both patients and healthy control groups. In patients, the mean ISR increased from 1.39 (95% CI: 1.13−1.65) at baseline to 2.48 (1.93−3.03) and 3.73 (3.13−4.38) following the first and second dosages, respectively. In multivariate analysis, the higher count of lymphocytes [OR: 8.57 (95% CI: 1.51−48.75); p = 0.02] and history of obtaining COVID-19 infection before transplantation [OR: 6.24 (95% CI: 1.17−33.15); p = 0.03] remained the predictors of the stronger immune response following two doses of the RBD-TT conjugated vaccine. Moreover, we found that the immunogenicity of the COVID-19 vaccine shortly after transplantation could be influenced by pre-transplant COVID-19 vaccination. Interpretation: The RBD-TT conjugated SARS-CoV-2 vaccine was safe, highly immunogenic, and affordable early after autologous transplants. Funding: This work was mainly financed by the Hematology-Oncology-Stem Cell Transplantation Research Center (HORCSCT) of Tehran University and the Pasteur Institute of Iran.
Collapse
|
30
|
Barkhordar M, Chahardouli B, Biglari A, Ahmadvand M, Bahri T, Alaeddini F, Sharifi Aliabadi L, Noorani SS, Bagheri Amiri F, Biglari M, Shemshadi MR, Ghavamzadeh A, Vaezi M. Three doses of a recombinant conjugated SARS-CoV-2 vaccine early after allogeneic hematopoietic stem cell transplantation: predicting indicators of a high serologic response-a prospective, single-arm study. Front Immunol 2023; 14:1169666. [PMID: 37153556 PMCID: PMC10154585 DOI: 10.3389/fimmu.2023.1169666] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 03/24/2023] [Indexed: 05/09/2023] Open
Abstract
Background Allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients must be vaccinated against SARS-CoV-2 as quickly as possible after transplantation. The difficulty in obtaining recommended SARS-CoV-2 vaccines for allo-HSCT recipients motivated us to utilize an accessible and affordable SARS-CoV-2 vaccine with a recombinant receptor-binding domain (RBD)-tetanus toxoid (TT)-conjugated platform shortly after allo-HSCT in the developing country of Iran. Methods This prospective, single-arm study aimed to investigate immunogenicity and its predictors following a three-dose SARS-CoV-2 RBD-TT-conjugated vaccine regimen administered at 4-week (± 1-week) intervals in patients within 3-12 months post allo-HSCT. An immune status ratio (ISR) was measured at baseline and 4 weeks (± 1 week) after each vaccine dose using a semiquantitative immunoassay. Using the median ISR as a cut-off point for immune response intensity, we performed a logistic regression analysis to determine the predictive impact of several baseline factors on the intensity of the serologic response following the third vaccination dose. Results Thirty-six allo-HSCT recipients, with a mean age of 42.42 years and a median time of 133 days between hematopoietic stem cell transplant (allo-HSCT) and the start of vaccination, were analyzed. Our findings, using the generalized estimating equation (GEE) model, indicated that, compared with the baseline ISR of 1.55 [95% confidence interval (CI) 0.94 to 2.17], the ISR increased significantly during the three-dose SARS-CoV-2 vaccination regimen. The ISR reached 2.32 (95% CI 1.84 to 2.79; p = 0.010) after the second dose and 3.87 (95% CI 3.25 to 4.48; p = 0.001) after the third dose of vaccine, reflecting 69.44% and 91.66% seropositivity, respectively. In a multivariate logistic regression analysis, the female sex of the donor [odds ratio (OR) 8.67; p = 0.028] and a higher level donor ISR at allo-HSCT (OR 3.56; p = 0.050) were the two positive predictors of strong immune response following the third vaccine dose. No serious adverse events (i.e., grades 3 and 4) were observed following the vaccination regimen. Conclusions We concluded that early vaccination of allo-HSCT recipients with a three-dose RBD-TT-conjugated SARS-CoV-2 vaccine is safe and could improve the early post-allo-HSCT immune response. We further believe that the pre-allo-HSCT SARS-CoV-2 immunization of donors may enhance post-allo-HSCT seroconversion in allo-HSCT recipients who receive the entire course of the SARS-CoV-2 vaccine during the first year after allo-HSCT.
Collapse
Affiliation(s)
- Maryam Barkhordar
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Mohammad Vaezi, ; Maryam Barkhordar,
| | - Bahram Chahardouli
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Biglari
- Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ahmadvand
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tanaz Bahri
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Alaeddini
- Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Leyla Sharifi Aliabadi
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seied Saeid Noorani
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Bagheri Amiri
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Biglari
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Shemshadi
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Cancer & Cell Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Mohammad Vaezi, ; Maryam Barkhordar,
| |
Collapse
|
31
|
COVID-19 Vaccination Response and Its Practical Application in Patients With Chronic Lymphocytic Leukemia. Hemasphere 2023; 7:e811. [PMID: 36570695 PMCID: PMC9771252 DOI: 10.1097/hs9.0000000000000811] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/09/2022] [Indexed: 12/27/2022] Open
Abstract
Patients with chronic lymphocyticleukemia (CLL) typically have innate/adaptive immune system dysregulation, thus the protective effect of coronavirus disease 2019 (COVID-19) vaccination remains uncertain. This prospective review evaluates vaccination response in these patients, including seropositivity rates by CLL treatment status, type of treatment received, and timing of vaccination. Antibody persistence, predictors of poor vaccine response, and severity of COVID-19 infection in vaccinated patients were also analyzed. Practical advice on the clinical management of patients with CLL is provided. Articles reporting COVID-19 vaccination in patients with CLL, published January 1, 2021-May 1, 2022, were included. Patients with CLL displayed the lowest vaccination responses among hematologic malignancies; however, seropositivity increased with each vaccination. One of the most commonly reported independent risk factors for poor vaccine response was active CLL treatment; others included hypogammaglobulinemia and age >65-70 years. Patients who were treatment-naive, off therapy, in remission, or who had a prior COVID-19 infection displayed the greatest responses. Further data are needed on breakthrough infection rates and a heterologous booster approach in patients with hematologic malignancies. Although vaccine response was poor for patients on active therapy regardless of treatment type, CLL management in the context of COVID-19 should aim to avoid delays in antileukemic treatment, especially with the advent of numerous strategies to mitigate risk of severe COVID-19 such as pre-exposure prophylaxis, and highly effective antivirals and monoclonal antibody therapy upon confirmed infection. Patients with CLL should remain vigilant in retaining standard prevention measures such as masks, social distancing, and hand hygiene.
Collapse
|
32
|
Uaprasert N, Pitakkitnukun P, Tangcheewinsirikul N, Chiasakul T, Rojnuckarin P. Immunogenicity and risks associated with impaired immune responses following SARS-CoV-2 vaccination and booster in hematologic malignancy patients: an updated meta-analysis. Blood Cancer J 2022; 12:173. [PMID: 36550105 PMCID: PMC9780106 DOI: 10.1038/s41408-022-00776-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Patients with hematologic malignancies (HM) have demonstrated impaired immune responses following SARS-CoV-2 vaccination. Factors associated with poor immunogenicity remain largely undetermined. A literature search was conducted using PubMed, EMBASE, Cochrane, and medRxiv databases to identify studies that reported humoral or cellular immune responses (CIR) following complete SARS-CoV-2 vaccination. The primary aim was to estimate the seroconversion rate (SR) following complete SARS-CoV-2 vaccination across various subtypes of HM diseases and treatments. The secondary aims were to determine the rates of development of neutralizing antibodies (NAb) and CIR following complete vaccination and SR following booster doses. A total of 170 studies were included for qualitative and quantitative analysis of primary and secondary outcomes. A meta-analysis of 150 studies including 20,922 HM patients revealed a pooled SR following SARS-CoV-2 vaccination of 67.7% (95% confidence interval [CI], 64.8-70.4%; I2 = 94%). Meta-regression analysis showed that patients with lymphoid malignancies, but not myeloid malignancies, had lower seroconversion rates than those with solid cancers (R2 = 0.52, P < 0.0001). Patients receiving chimeric antigen receptor T-cells (CART), B-cell targeted therapies or JAK inhibitors were associated with poor seroconversion (R2 = 0.39, P < 0.0001). The pooled NAb and CIR rates were 52.8% (95% CI; 45.8-59.7%, I2 = 87%) and 66.6% (95% CI, 57.1-74.9%; I2 = 86%), respectively. Approximately 20.9% (95% CI, 11.4-35.1%, I2 = 90%) of HM patients failed to elicit humoral and cellular immunity. Among non-seroconverted patients after primary vaccination, only 40.5% (95% CI, 33.0-48.4%; I2 = 87%) mounted seroconversion after the booster. In conclusion, HM patients, especially those with lymphoid malignancies and/or receiving CART, B-cell targeted therapies, or JAK inhibitors, showed poor SR after SARS-CoV-2 vaccination. A minority of patients attained seroconversion after booster vaccination. Strategies to improve immune response in these severely immunosuppressed patients are needed.
Collapse
Affiliation(s)
- Noppacharn Uaprasert
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand.
- Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand.
| | - Palada Pitakkitnukun
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nuanrat Tangcheewinsirikul
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Thita Chiasakul
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Ponlapat Rojnuckarin
- Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
33
|
Low Spike Antibody Levels and Impaired BA.4/5 Neutralization in Patients with Multiple Myeloma or Waldenstrom's Macroglobulinemia after BNT162b2 Booster Vaccination. Cancers (Basel) 2022; 14:cancers14235816. [PMID: 36497296 PMCID: PMC9737406 DOI: 10.3390/cancers14235816] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Patients with symptomatic monoclonal gammopathies have impaired humoral responses to COVID-19 vaccination. Their ability to recognize SARS-CoV-2 Omicron variants is of concern. We compared the response to BNT162b2 mRNA vaccinations of patients with multiple myeloma (MM, n = 60) or Waldenstrom's macroglobulinemia (WM, n = 20) with healthy vaccine recipients (n = 37). Patient cohorts on active therapy affecting B cell development had impaired binding and neutralizing antibody (NAb) response rate and magnitude, including several patients lacking responses, even after a 3rd vaccine dose, whereas non-B cell depleting therapies had a lesser effect. In contrast, MM and WM cohorts off-therapy showed increased NAb with a broad response range. ELISA Spike-Receptor Binding Domain (RBD) Ab titers in healthy vaccine recipients and patient cohorts were good predictors of the ability to neutralize not only the original WA1 but also the most divergent Omicron variants BA.4/5. Compared to WA1, significantly lower NAb responses to BA.4/5 were found in all patient cohorts on-therapy. In contrast, the MM and WM cohorts off-therapy showed a higher probability to neutralize BA.4/5 after the 3rd vaccination. Overall, the boost in NAb after the 3rd dose suggests that repeat vaccination of MM and WM patients is beneficial even under active therapy.
Collapse
|
34
|
Chen Z, Zhu P, Liu Z, Zhu B, Yin G, Ming J, Song R, Pan Q, Li T, Jiang X, Wang B, Liu S, Cai H, Wang J, Han Y, Lin Z, Hong Y, Chen M, Peng M, Hu L, Cai D, Ren H. Weakened humoral immune responses of inactivated SARS-CoV-2 vaccines in patients with solid tumors. Cancer Commun (Lond) 2022; 43:280-284. [PMID: 36330800 PMCID: PMC9877898 DOI: 10.1002/cac2.12368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/24/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Zhiwei Chen
- Department of Infectious DiseasesKey Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral Hepatitisthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Peng Zhu
- Department of Gastrointestinal Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Zuojin Liu
- Department of Hepatobiliary Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Bin Zhu
- Department of Thoracic and Cardiovascular Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Guobing Yin
- Department of Breast and Thyroid Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Jia Ming
- Department of Breast and Thyroid Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Rui Song
- Department of Infectious DiseasesKey Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral Hepatitisthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Qingbo Pan
- Department of Infectious DiseasesKey Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral Hepatitisthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Tong Li
- Department of Gastrointestinal Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xingwei Jiang
- Department of Gynaecology and ObstetricsReproductive Medicine Centerthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Bing Wang
- Department of Thoracic and Cardiovascular Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Shuaibin Liu
- Department of Gynaecology and ObstetricsReproductive Medicine Centerthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hongxing Cai
- Department of Gastrointestinal Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Jingjie Wang
- Department of Gastrointestinal Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Yuling Han
- Department of Breast and Thyroid Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Zijing Lin
- Department of Breast and Thyroid Surgerythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Yang Hong
- Department of Gynaecology and ObstetricsReproductive Medicine Centerthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Min Chen
- Department of Infectious DiseasesKey Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral Hepatitisthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Mingli Peng
- Department of Infectious DiseasesKey Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral Hepatitisthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Lina Hu
- Department of Gynaecology and ObstetricsReproductive Medicine Centerthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Dachuan Cai
- Department of Infectious DiseasesKey Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral Hepatitisthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Hong Ren
- Department of Infectious DiseasesKey Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education)Institute for Viral Hepatitisthe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| |
Collapse
|
35
|
Meeuwes FO, Brink M, van der Poel MWM, Kersten MJ, Wondergem M, Mutsaers PGNJ, Böhmer L, Woei-A-Jin S, Visser O, Oostvogels R, Jansen PM, Diepstra A, Snijders TJF, Plattel WJ, Huls GA, Vermaat JSP, Nijland M. Impact of rituximab on treatment outcomes of patients with angioimmunoblastic T-cell lymphoma; a population-based analysis. Eur J Cancer 2022; 176:100-109. [PMID: 36208568 DOI: 10.1016/j.ejca.2022.09.008] [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: 06/17/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with angioimmunoblastic T-cell lymphoma (AITL) are treated with cyclophosphamide, doxorubicin, vincristine and prednisone with or without etoposide (CHO(E)P). In the majority of cases, Epstein-Barr virus (EBV)-positive B-cells are present in the tumour. There is paucity of research examining the effect of rituximab when added to CHO(E)P. In this nationwide, population-based study, we analysed the impact of rituximab on overall response rate (ORR), progression-free survival (PFS) and overall survival (OS) of patients with AITL. METHODS Patients with AITL diagnosed between 2014 and 2020 treated with ≥one cycle of CHO(E)P with or without rituximab were identified in the Netherlands Cancer Registry. Survival follow-up was up to 1st February 2022. Baseline characteristics, best response during first-line treatment and survival were collected. PFS was defined as the time from diagnosis to relapse or to all-cause-death. OS was defined as the time from diagnosis to all-cause-death. Multivariable analysis for the risk of mortality was performed using Cox regression. FINDINGS Out of 335 patients, 146 patients (44%) received R-CHO(E)P. Rituximab was more frequently used in patients with a B-cell infiltrate (71% versus 89%, p < 0·01). The proportion of patients who received autologous stem cell transplantation (ASCT) was similar between CHO(E)P and R-CHO(E)P (27% versus 30%, respectively). The ORR and 2-year PFS for patients who received CHO(E)P and R-CHO(E)P were 71% and 78% (p = 0·01), and 40% and 45% (p = 0·12), respectively. The 5-year OS was 47% and 40% (p = 0·99), respectively. In multivariable analysis, IPI-score 3-5, no B-cell infiltrate and no ASCT were independent prognostic factors for risk of mortality, whereas the use of rituximab was not. INTERPRETATION Although the addition of rituximab to CHO(E)P improved ORR for patients with AITL, the PFS and OS did not improve.
Collapse
Affiliation(s)
- Frederik O Meeuwes
- Department of Hematology, Treant Hospital, Emmen, the Netherlands; Department of Hematology, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirian Brink
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands
| | - Marjolein W M van der Poel
- Department of Internal Medicine, Division of Hematology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marie José Kersten
- Department of Hematology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mariëlle Wondergem
- Department of Hematology, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Pim G N J Mutsaers
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Lara Böhmer
- Department of Hematology, Haga Hospital, The Hague, the Netherlands
| | - Sherida Woei-A-Jin
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Otto Visser
- Department of Hematology, Isala Hospital, Zwolle, the Netherlands
| | - Rimke Oostvogels
- Department of Hematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Patty M Jansen
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arjan Diepstra
- Department of Pathology, University Medical Center Groningen, Groningen, the Netherlands
| | - Tjeerd J F Snijders
- Department of Hematology, Medisch Spectrum Twente, Enschede, the Netherlands
| | - Wouter J Plattel
- Department of Hematology, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerwin A Huls
- Department of Hematology, University Medical Center Groningen, Groningen, the Netherlands
| | - Joost S P Vermaat
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marcel Nijland
- Department of Hematology, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
36
|
Haggenburg S, Hofsink Q, Rutten CE, Nijhof IS, Hazenberg MD, Goorhuis A. SARS-CoV-2 vaccine-induced humoral and cellular immunity in patients with hematologic malignancies. Semin Hematol 2022; 59:192-197. [PMID: 36805887 PMCID: PMC9674560 DOI: 10.1053/j.seminhematol.2022.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
Patients with hematologic conditions have a higher risk of severe COVID-19 and COVID-19-related death. This is related to immune deficiencies induced by hematologic conditions and/or the treatment thereof. Prospective vaccine immunogenicity studies have demonstrated that in the majority of patients, a 3-dose COVID-19 vaccination schedule leads to antibody concentrations comparable to levels obtained in healthy adults after a 2-dose schedule. In B cell depleted patients, humoral responses are poor, however vaccination did induce potent cellular immune responses. The effect of 3-dose vaccination schedules and COVID-19 booster vaccinations on the protection of patients with hematologic malignancies against severe COVID-19 and COVID-19 related death remains to be confirmed by population-based vaccine effectiveness studies.
Collapse
Affiliation(s)
- Sabine Haggenburg
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands; Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Quincy Hofsink
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| | - Caroline E. Rutten
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Inger S. Nijhof
- Department of Hematology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands,Department of Internal Medicine-Hematology, St. Antonius Hospital, Nieuwegein, The Netherlands,Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Mette D. Hazenberg
- Department of Hematology, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands,Amsterdam institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands,Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands,Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - Abraham Goorhuis
- Department of Infectious Diseases, Centre of Tropical Medicine and Travel Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Manjappa S, Phi HQ, Lee LW, Onstad L, Gill DB, Connelly-Smith L, Krakow EF, Flowers ME, Carpenter PA, Hill JA, Lee SJ. Humoral and Cellular Immune Response to Covid-19 Vaccination in Patients with Chronic Graft-versus-Host Disease on Immunosuppression. Transplant Cell Ther 2022; 28:784.e1-784.e9. [PMID: 36058550 PMCID: PMC9436787 DOI: 10.1016/j.jtct.2022.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022]
Abstract
Chronic graft-versus-host disease (cGVHD) and its management with immunosuppressive therapies increase the susceptibility to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as well as progression to severe Coronavirus 19 disease (COVID-19). Vaccination against COVID-19 is strongly recommended, but efficacy data are limited in this patient population. In this study, responses to COVID-19 vaccination were measured at 3 time points—after the initial vaccine series, before the third dose, and after the third dose—in adults with cGVHD receiving immunosuppressive therapy. Humoral response was measured by quantitative anti-spike antibody and neutralizing antibody levels. Anti-nucleocapsid antibody levels were measured to detect natural infection. T cell response was evaluated by a novel immunosequencing technique combined with immune repertoire profiling from cryopreserved peripheral blood mononuclear cell samples. Present or absent T cell responses were determined by the relative proportion of unique SARS-CoV-2-associated T cell receptor sequences (“breadth”) plus clonal expansion of the response (“depth”) compared with those in a reference population. Based on both neutralizing antibody and T cell responses, patients were categorized as vaccine responders (both detected), nonresponders (neither detected), or mixed (one but not both detected). Thirty-two patients were enrolled for the initial series, including 17 (53%) positive responders, 7 (22%) mixed responders, and 8 (25%) nonresponders. All but one patient categorized as mixed responders had humoral responses while lacking T cell responses. No statistical differences were observed in patient characteristics among the 3 groups of patients categorized by immune response, although sample sizes were limited. Significant positive correlations were observed between the robustness of cellular and humoral responses after the initial series. Among the 20 patients with paired samples (pre- and post-third dose), a third vaccination resulted in increased neutralizing antibody titers. cGVHD worsened in 10 patients (26%; 6 after the initial series and 4 after the third dose), necessitating escalation of immunosuppressive doses in 5 patients, although 4 had been tapering immunosuppression and 5 had already worsening cGVHD at the time of vaccination, and a clear association between COVID-19 vaccination and cGVHD could not be drawn. Among the patients with cGVHD on immunosuppressive therapy, 72% demonstrated a neutralizing antibody response after a 2-dose primary COVID-19 vaccination, two-thirds of whom also developed a T cell response; 25% had neither a humoral nor a T cell response. A third dose further amplified the antibody response.
Collapse
Affiliation(s)
- Shivaprasad Manjappa
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Huy Q Phi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Lik Wee Lee
- Adaptive Biotechnologies, Seattle, Washington
| | - Lynn Onstad
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Laura Connelly-Smith
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Elizabeth F Krakow
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Mary E Flowers
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington
| | - Joshua A Hill
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington; Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Stephanie J Lee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
38
|
Chung A, Banbury B, Vignali M, Huang CY, Asoori S, Johnson R, Kurtz T, Arora S, Wong SW, Shah N, Martin TG, Wolf JL. Antibody and T-cell responses by ultra-deep T-cell receptor immunosequencing after COVID-19 vaccination in patients with plasma cell dyscrasias. Br J Haematol 2022; 199:520-528. [PMID: 36041779 PMCID: PMC9538250 DOI: 10.1111/bjh.18434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022]
Abstract
We investigated antibody and coronavirus disease 2019 (COVID‐19)‐specific T‐cell mediated responses via ultra‐deep immunosequencing of the T‐cell receptor (TCR) repertoire in patients with plasma cell dyscrasias (PCD). We identified 364 patients with PCD who underwent spike antibody testing using commercially available spike‐receptor binding domain immunoglobulin G antibodies ≥2 weeks after completion of the initial two doses of mRNA vaccines or one dose of JNJ‐78436735. A total of 56 patients underwent TCR immunosequencing after vaccination. Overall, 86% tested within 6 months of vaccination had detectable spike antibodies. Increasing age, use of anti‐CD38 or anti‐B‐cell maturation antigen therapy, and receipt of BNT162b2 (vs. mRNA‐1273) were associated with lower antibody titres. We observed an increased proportion of TCRs associated with surface glycoprotein regions of the COVID‐19 genome after vaccination, consistent with spike‐specific T‐cell responses. The median spike‐specific T‐cell breadth was 3.11 × 10−5, comparable to those in healthy populations after vaccination. Although spike‐specific T‐cell breadth correlated with antibody titres, patients without antibody responses also demonstrated spike‐specific T‐cell responses. Patients receiving mRNA‐1273 had higher median spike‐specific T‐cell breadth than those receiving BNT162b2 (p = 0.01). Although patients with PCD are often immunocompromised due to underlying disease and treatments, COVID‐19 vaccination can still elicit humoral and T‐cell responses and remain an important intervention in this patient population.
Collapse
Affiliation(s)
- Alfred Chung
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | | | | | - Chiung-Yu Huang
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Sireesha Asoori
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | - Rachel Johnson
- American University of the Caribbean School of Medicine, St. Maarten
| | - Theodore Kurtz
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Shagun Arora
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | - Sandy W Wong
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | - Nina Shah
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | - Thomas G Martin
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey L Wolf
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
39
|
Haggenburg S, Hofsink Q, Lissenberg-Witte BI, Broers AEC, van Doesum JA, van Binnendijk RS, den Hartog G, Bhoekhan MS, Haverkate NJE, Burger JA, Bouhuijs JH, Smits GP, Wouters D, van Leeuwen EMM, Bontkes HJ, Kootstra NA, Zweegman S, Kater AP, Heemskerk MHM, Groen K, van Meerten T, Mutsaers PGNJ, Beaumont T, van Gils MJ, Goorhuis A, Rutten CE, Hazenberg MD, Nijhof IS. Antibody Response in Immunocompromised Patients With Hematologic Cancers Who Received a 3-Dose mRNA-1273 Vaccination Schedule for COVID-19. JAMA Oncol 2022; 8:1477-1483. [PMID: 35951338 DOI: 10.1001/jamaoncol.2022.3227] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Importance It has become common practice to offer immunocompromised patients with hematologic cancers a third COVID-19 vaccination dose, but data substantiating this are scarce. Objective To assess whether a third mRNA-1273 vaccination is associated with increased neutralizing antibody concentrations in immunocompromised patients with hematologic cancers comparable to levels obtained in healthy individuals after the standard 2-dose mRNA-1273 vaccination schedule. Design, Setting, and Participants This prospective observational cohort study was conducted at 4 university hospitals in the Netherlands and included 584 evaluable patients spanning the spectrum of hematologic cancers and 44 randomly selected age-matched adults without malignant or immunodeficient comorbidities. Exposures One additional mRNA-1273 vaccination 5 months after completion of the standard 2-dose mRNA-1273 vaccination schedule. Main Outcomes and Measures Serum immunoglobulin G (IgG) antibodies to spike subunit 1 (S1) antigens prior to and 4 weeks after a third mRNA-1273 vaccination, and antibody neutralization capacity of wild-type, Delta, and Omicron variants in a subgroup of patients. Results In this cohort of 584 immunocompromised patients with hematologic cancers (mean [SD] age, 60 [11.2] years; 216 [37.0%] women), a third mRNA-1273 vaccination was associated with median S1-IgG concentrations comparable to concentrations obtained by healthy individuals after the 2-dose mRNA-1273 schedule. The rise in S1-IgG concentration after the third vaccination was most pronounced in patients with a recovering immune system, but potent responses were also observed in patients with persistent immunodeficiencies. Specifically, patients with myeloid cancers or multiple myeloma and recipients of autologous or allogeneic hematopoietic cell transplantation (HCT) reached median S1-IgG concentrations similar to those obtained by healthy individuals after a 2-dose schedule. Patients receiving or shortly after completing anti-CD20 therapy, CD19-directed chimeric antigen receptor T-cell therapy recipients, and patients with chronic lymphocytic leukemia receiving ibrutinib were less responsive or unresponsive to the third vaccination. In the 27 patients who received cell therapy between the second and third vaccination, S1 antibodies were preserved, but a third mRNA-1273 vaccination was not associated with significantly enhanced S1-IgG concentrations except for patients with multiple myeloma receiving autologous HCT. A third vaccination was associated with significantly improved neutralization capacity per antibody. Conclusions and Relevance Results of this cohort study support that the primary schedule for immunocompromised patients with hematologic cancers should be supplemented with a delayed third vaccination. Patients with B-cell lymphoma and allogeneic HCT recipients need to be revaccinated after treatment or transplantation. Trial Registration EudraCT Identifier: 2021-001072-41.
Collapse
Affiliation(s)
- Sabine Haggenburg
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands
| | - Quincy Hofsink
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands
| | - Birgit I Lissenberg-Witte
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Annoek E C Broers
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Jaap A van Doesum
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rob S van Binnendijk
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Gerco den Hartog
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Michel S Bhoekhan
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands
| | - Nienke J E Haverkate
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Judith A Burger
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Joey H Bouhuijs
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Gaby P Smits
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Dorine Wouters
- Central Diagnostic Laboratory, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ester M M van Leeuwen
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hetty J Bontkes
- Laboratory Medical Immunology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Neeltje A Kootstra
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Arnon P Kater
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Kaz Groen
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands
| | - Tom van Meerten
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pim G N J Mutsaers
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Tim Beaumont
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marit J van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Abraham Goorhuis
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Caroline E Rutten
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Mette D Hazenberg
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.,Department of Hematopoiesis, Sanquin Research, Amsterdam, the Netherlands
| | - Inger S Nijhof
- Department of Hematology, Amsterdam UMC, Vrije Universiteit, Amsterdam, the Netherlands.,Department of Internal Medicine-Hematology, St Antonius Hospital, Nieuwegein, the Netherlands
| | | |
Collapse
|
40
|
Huygens S, Hofsink Q, Nijhof IS, Goorhuis A, Kater AP, te Boekhorst PAW, Swaneveld F, Novotný VMJ, Bogers S, Welkers MRA, Papageorgiou G, Rijnders BJ, Heijmans J. Hyperimmune Globulin for Severely Immunocompromised Patients Hospitalized With Coronavirus Disease 2019: A Randomized, Controlled Trial. J Infect Dis 2022; 227:206-210. [PMID: 35921542 PMCID: PMC9384649 DOI: 10.1093/infdis/jiac334] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The aim of this randomized, controlled trial is to determine whether antisevere acute respiratory syndrome coronavirus 2 hyperimmune globulin (COVIG) protects against severe coronavirus disease 2019 (COVID-19) in severely immunocompromised, hospitalized, COVID-19 patients. METHODS Patients were randomly assigned to receive COVIG or intravenous immunoglobulin (IVIG) without SARS-CoV-2 antibodies. RESULTS Severe COVID-19 was observed in 2 of 10 (20%) patients treated with COVIG compared to 7 of 8 (88%) in the IVIG control group (P = .015, Fisher's exact test). CONCLUSIONS Antisevere acute respiratory syndrome coronavirus 2 hyperimmune globulin may be a valuable treatment in severely immunocompromised, hospitalized, COVID-19 patients and should be considered when no monoclonal antibody therapies are available.
Collapse
Affiliation(s)
| | | | - Inger S Nijhof
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands,Department of Internal Medicine-Hematology, St. Antonius Hospital, Nieuwegein, The Netherlands
| | - Abraham Goorhuis
- Department of Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Arnon P Kater
- Department of Hematology, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Francis Swaneveld
- Unit of Transfusion Medicine, Sanquin Blood Supply Foundation, Amsterdam, The Netherlands
| | - Věra M J Novotný
- Unit of Transfusion Medicine, Sanquin Blood Supply Foundation, Amsterdam, The Netherlands
| | - Susanne Bogers
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Matthijs R A Welkers
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | | | - Bart J Rijnders
- Department of Internal Medicine, Section of Infectious Diseases, and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jarom Heijmans
- Correspondence: J. Heijmans, MD, PhD, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ()
| |
Collapse
|
41
|
Huygens S, Oude Munnink B, Gharbharan A, Koopmans M, Rijnders B. Sotrovimab Resistance and Viral Persistence After Treatment of Immunocompromised Patients Infected With the Severe Acute Respiratory Syndrome Coronavirus 2 Omicron Variant. Clin Infect Dis 2022; 76:e507-e509. [PMID: 35867699 PMCID: PMC9384506 DOI: 10.1093/cid/ciac601] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Viral evolution was evaluated in 47 immunocompromised patients treated with sotrovimab. Sequencing of SARS-CoV-2 following therapy was successful in 16. Mutations associated with sotrovimab resistance were documented in 6; viral replication continued after 30 days in 5. Combination antibody therapy may be required to avoid acquired resistance in immunocompromised patients.
Collapse
Affiliation(s)
| | | | - Arvind Gharbharan
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, University Medical Center, Rotterdam, The Netherlands
| | | | - Bart Rijnders
- Correspondence: B. J. A. Rijnders, Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Dr. Molewaterplein 40 3015 GD, Rotterdam, The Netherlands ()
| |
Collapse
|
42
|
Green NS, Van Doren L, Licursi M, Billings DD, Sandoval LA, Feit YMZ, Hod EA. Anti-SARS-CoV-19 antibodies in children and adults with sickle cell disease: A single-site analysis in New York City. Br J Haematol 2022; 198:680-683. [PMID: 35759223 PMCID: PMC9350182 DOI: 10.1111/bjh.18294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Nancy S Green
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplantation, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA.,Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Layla Van Doren
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Maureen Licursi
- Department of Pediatrics, Division of Hematology, Oncology and Stem Cell Transplantation, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Daniel D Billings
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Luke A Sandoval
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Yona M Z Feit
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| | - Eldad A Hod
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center - New York Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
43
|
Piñana JL, López-Corral L, Martino R, Vazquez L, Pérez A, Martin-Martin G, Gago B, Sanz-Linares G, Sanchez-Salinas A, Villalon L, Conesa-Garcia V, Olave MT, Corona M, Marcos-Corrales S, Tormo M, Hernández-Rivas JÁ, Montoro J, Rodriguez-Fernandez A, Risco-Gálvez I, Rodríguez-Belenguer P, Hernandez-Boluda JC, García-Cadenas I, Ruiz-García M, Muñoz-Bellido JL, Solano C, Cedillo Á, Sureda A, Navarro D. SARS-CoV-2 vaccine response and rate of breakthrough infection in patients with hematological disorders. J Hematol Oncol 2022; 15:54. [PMID: 35526045 PMCID: PMC9077637 DOI: 10.1186/s13045-022-01275-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Background The clinical efficacy of SARS-CoV-2 vaccines according to antibody response in immunosuppressed patients such as hematological patients has not yet been established. Patients and methods A prospective multicenter registry-based cohort study conducted from December 2020 to December 2021 by the Spanish transplant and cell therapy group was used to analyze the relationship of antibody response at 3–6 weeks after full vaccination (2 doses) with breakthrough SARS-CoV-2 infection in 1394 patients with hematological disorders. Results At a median follow-up of 165 days after complete immunization, 37 out of 1394 (2.6%) developed breakthrough SARS-CoV-2 infection at median of 77 days (range 7–195) after full vaccination. The incidence rate was 6.39 per 100 persons-year. Most patients were asymptomatic (19/37, 51.4%), whereas only 19% developed pneumonia. The mortality rate was 8%. Lack of detectable antibodies at 3–6 weeks after full vaccination was the only variable associated with breakthrough infection in multivariate logistic regression analysis (Odds Ratio 2.35, 95% confidence interval 1.2–4.6, p = 0.012). Median antibody titers were lower in cases than in non-cases [1.83 binding antibody units (BAU)/mL (range 0–4854.93) vs 730.81 BAU/mL (range 0–56,800), respectively (p = 0.007)]. We identified 250 BAU/mL as a cutoff above which incidence and severity of the infection were significantly lower. Conclusions Our study highlights the benefit of developing an antibody response in these highly immunosuppressed patients. Level of antibody titers at 3 to 6 weeks after 2-dose vaccination links with protection against both breakthrough infection and severe disease for non-Omicron SARS-CoV-2 variants. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01275-7.
Collapse
Affiliation(s)
- José Luis Piñana
- Division of Clinical Hematology, Hematology Department, Hospital Clínico Universitario de Valencia, Avda Blasco Ibañez, 17, 46010, Valencia, Spain. .,Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain.
| | - Lucia López-Corral
- Hematology Division, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Rodrigo Martino
- Hematology Division, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Lourdes Vazquez
- Hematology Division, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Ariadna Pérez
- Division of Clinical Hematology, Hematology Department, Hospital Clínico Universitario de Valencia, Avda Blasco Ibañez, 17, 46010, Valencia, Spain.,Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | - Beatriz Gago
- Hematology Division, Hospital Regional Universitario Carlos Haya, Malaga, Spain
| | - Gabriela Sanz-Linares
- Hematology Division, Institut Català Oncologia-Hospital Duran i Reynals, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Andrés Sanchez-Salinas
- Hematology Division, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Lucia Villalon
- Hematology Division, Hospital Universitario Fundación Alcorcón, Madrid, Spain
| | | | - María T Olave
- Hematology Division, Hospital Clínico Universitario Lozano Blesa, IIS Aragon, Saragossa, Spain
| | | | | | - Mar Tormo
- Division of Clinical Hematology, Hematology Department, Hospital Clínico Universitario de Valencia, Avda Blasco Ibañez, 17, 46010, Valencia, Spain.,Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | - Juan Montoro
- Hematology Division, Hospital universitario y politécnico La Fe, Valencia, Spain
| | | | | | - Pablo Rodríguez-Belenguer
- Research Program on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences, Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra, Barcelona, Spain
| | - Juan Carlos Hernandez-Boluda
- Division of Clinical Hematology, Hematology Department, Hospital Clínico Universitario de Valencia, Avda Blasco Ibañez, 17, 46010, Valencia, Spain.,Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain.,Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | | | | | | | - Carlos Solano
- Division of Clinical Hematology, Hematology Department, Hospital Clínico Universitario de Valencia, Avda Blasco Ibañez, 17, 46010, Valencia, Spain.,Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain.,Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - Ángel Cedillo
- Hematopoietic Stem Cell Transplantation and Cell Therapy Group (GETH) Office, Madrid, Spain
| | - Anna Sureda
- Hematology Division, Institut Català Oncologia-Hospital Duran i Reynals, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - David Navarro
- Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain.,Hospital Clinico Universitario de Valencia, Valencia, Spain
| | | |
Collapse
|
44
|
Grigg A. Living Long and Well After an Autograft. J Clin Oncol 2022; 40:1971-1975. [PMID: 35512253 DOI: 10.1200/jco.22.00302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Andrew Grigg
- Department Clinical Haematology, Austin Hospital, Heidelberg, Melbourne, Australia
| |
Collapse
|