1
|
Hua T, Fan R, Fan Y, Chen F. Immune response of COVID-19 vaccines in solid cancer patients: A meta-analysis. Hum Vaccin Immunother 2024; 20:2357424. [PMID: 38785118 PMCID: PMC11135846 DOI: 10.1080/21645515.2024.2357424] [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: 02/15/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
Solid cancer patients, compared to their healthy counterparts, are at a greater risk of contracting and suffering from severe complications and poorer prognosis after COVID-19 infections. They also have different immune responses after doses of COVID-19 vaccination, but limited evidence is available to reveal the effectiveness and help to guide immunization programs for this subpopulation; MEDLINE, Embase, Web of Science, Cochrane Library databases, and clinicaltrials.gov were used to search literature. The pooled seroconversion rate was calculated using a random-effects model and reported with a 95% confidence interval (CI); The review includes 66 studies containing serological responses after COVID-19 vaccination in 13,050 solid cancer patients and 8550 healthy controls. The pooled seropositive rates after the first dose in patients with solid cancer and healthy controls are 55.2% (95% CI 45.9%-64.5% N = 18) and 90.2% (95% CI 80.9%-96.6% N = 13), respectively. The seropositive rates after the second dose in patients with solid cancer and healthy controls are 87.6% (95% CI 84.1%-90.7% N = 50) and 98.9% (95% CI 97.6%-99.7% N = 35), respectively. The seropositive rates after the third dose in patients with solid cancer and healthy controls are 91.4% (95% CI 85.4%-95.9% N = 21) and 99.8% (95% CI 98.1%-100.0% N = 4), respectively. Subgroup analysis finds that study sample size, timing of antibody testing, and vaccine type have influence on the results; Seroconversion rates after COVID-19 vaccination are significantly lower in patients with solid malignancies, especially after the first dose, then shrinking gradually after the following two vaccinations, indicating that subsequent doses or a booster dose should be considered for the effectiveness of this subpopulation.
Collapse
Affiliation(s)
- Tiantian Hua
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Ru Fan
- Medical Statistics and Analysis Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yang Fan
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
| | - Feng Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, China
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Wu L, Yang L, Qian X, Hu W, Wang S, Yan J. Mannan-Decorated Lipid Calcium Phosphate Nanoparticle Vaccine Increased the Antitumor Immune Response by Modulating the Tumor Microenvironment. J Funct Biomater 2024; 15:229. [PMID: 39194667 DOI: 10.3390/jfb15080229] [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: 05/23/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
With the rapid development of tumor immunotherapy, nanoparticle vaccines have attracted much attention as potential therapeutic strategies. A systematic review and analysis must be carried out to investigate the effect of mannose modification on the immune response to nanoparticles in regulating the tumor microenvironment, as well as to explore its potential clinical application in tumor therapy. Despite the potential advantages of nanoparticle vaccines in immunotherapy, achieving an effective immune response in the tumor microenvironment remains a challenge. Tumor immune escape and the overexpression of immunosuppressive factors limit its clinical application. Therefore, our review explored how to intervene in the immunosuppressive mechanism in the tumor microenvironment through the use of mannan-decorated lipid calcium phosphate nanoparticle vaccines to improve the efficacy of immunotherapy in patients with tumors and to provide new ideas and strategies for the field of tumor therapy.
Collapse
Affiliation(s)
- Liusheng Wu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 19077, Singapore
| | - Lei Yang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xinye Qian
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wang Hu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shuang Wang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
3
|
Tobias J, Steinberger P, Wilkinson J, Klais G, Kundi M, Wiedermann U. SARS-CoV-2 Vaccines: The Advantage of Mucosal Vaccine Delivery and Local Immunity. Vaccines (Basel) 2024; 12:795. [PMID: 39066432 PMCID: PMC11281395 DOI: 10.3390/vaccines12070795] [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: 06/12/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Immunity against respiratory pathogens is often short-term, and, consequently, there is an unmet need for the effective prevention of such infections. One such infectious disease is coronavirus disease 19 (COVID-19), which is caused by the novel Beta coronavirus SARS-CoV-2 that emerged around the end of 2019. The World Health Organization declared the illness a pandemic on 11 March 2020, and since then it has killed or sickened millions of people globally. The development of COVID-19 systemic vaccines, which impressively led to a significant reduction in disease severity, hospitalization, and mortality, contained the pandemic's expansion. However, these vaccines have not been able to stop the virus from spreading because of the restricted development of mucosal immunity. As a result, breakthrough infections have frequently occurred, and new strains of the virus have been emerging. Furthermore, SARS-CoV-2 will likely continue to circulate and, like the influenza virus, co-exist with humans. The upper respiratory tract and nasal cavity are the primary sites of SARS-CoV-2 infection and, thus, a mucosal/nasal vaccination to induce a mucosal response and stop the virus' transmission is warranted. In this review, we present the status of the systemic vaccines, both the approved mucosal vaccines and those under evaluation in clinical trials. Furthermore, we present our approach of a B-cell peptide-based vaccination applied by a prime-boost schedule to elicit both systemic and mucosal immunity.
Collapse
Affiliation(s)
- Joshua Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Joy Wilkinson
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gloria Klais
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Kundi
- Department of Environmental Health, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
4
|
Goodyear CS, Patel A, Barnes E, Willicombe M, Siebert S, de Silva TI, Snowden JA, Lim SH, Bowden SJ, Billingham L, Richter A, Carroll M, Carr EJ, Beale R, Rea D, Parry H, Pirrie S, Lim Z, Satsangi J, Dunachie SJ, Cook G, Miller P, Basu N, Gilmour A, Hodgkins AM, Evans L, Hughes A, Longet S, Meacham G, Yong KL, A'Hearne MJ, Koh MBC, Burns SO, Orchard K, Paterson C, McIlroy G, Murray SM, Thomson T, Dimitriadis S, Goulston L, Miller S, Keillor V, Prendecki M, Thomas D, Kirkham A, McInnes IB, Kearns P. Immunogenicity of third dose COVID-19 vaccine strategies in patients who are immunocompromised with suboptimal immunity following two doses (OCTAVE-DUO): an open-label, multicentre, randomised, controlled, phase 3 trial. THE LANCET. RHEUMATOLOGY 2024; 6:e339-e351. [PMID: 38734019 DOI: 10.1016/s2665-9913(24)00065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The humoral and T-cell responses to booster COVID-19 vaccine types in multidisease immunocompromised individuals who do not generate adequate antibody responses to two COVID-19 vaccine doses, is not fully understood. The OCTAVE DUO trial aimed to determine the value of third vaccinations in a wide range of patients with primary and secondary immunodeficiencies. METHODS OCTAVE-DUO was a prospective, open-label, multicentre, randomised, controlled, phase 3 trial investigating humoral and T-cell responses in patients who are immunocompromised following a third vaccine dose with BNT162b2 or mRNA-1273, and of NVX-CoV2373 for those with lymphoid malignancies. We recruited patients who were immunocompromised from 11 UK hospitals, aged at least 18 years, with previous sub-optimal responses to two doses of SARS-CoV-2 vaccine. Participants were randomly assigned 1:1 (1:1:1 for those with lymphoid malignancies), stratified by disease, previous vaccination type, and anti-spike antibody response following two doses. Individuals with lived experience of immune susceptibility were involved in the study design and implementation. The primary outcome was vaccine-specific immunity defined by anti-SARS-CoV-2 spike antibodies (Roche Diagnostics UK and Ireland, Burgess Hill, UK) and T-cell responses (Oxford Immunotec, Abingdon, UK) before and 21 days after the third vaccine dose analysed by a modified intention-to-treat analysis. The trial is registered with the ISRCTN registry, ISRCTN 15354495, and the EU Clinical Trials Register, EudraCT 2021-003632-87, and is complete. FINDINGS Between Aug 4, 2021 and Mar 31, 2022, 804 participants across nine disease cohorts were randomly assigned to receive BNT162b2 (n=377), mRNA-1273 (n=374), or NVX-CoV2373 (n=53). 356 (45%) of 789 participants were women, 433 (55%) were men, and 659 (85%) of 775 were White. Anti-SARS-CoV-2 spike antibodies measured 21 days after the third vaccine dose were significantly higher than baseline pre-third dose titres in the modified intention-to-treat analysis (median 1384 arbitrary units [AU]/mL [IQR 4·3-7990·0] compared with median 11·5 AU/mL [0·4-63·1]; p<0·001). Of participants who were baseline low responders, 380 (90%) of 423 increased their antibody concentrations to more than 400 AU/mL. Conversely, 166 (54%) of 308 baseline non-responders had no response after the third dose. Detectable T-cell responses following the third vaccine dose were seen in 494 (80%) of 616 participants. There were 24 serious adverse events (BNT612b2 eight [33%] of 24, mRNA-1273 12 [50%], NVX-CoV2373 four [17%]), two (8%) of which were categorised as vaccine-related. There were seven deaths (1%) during the trial, none of which were vaccine-related. INTERPRETATION A third vaccine dose improved the serological and T-cell response in the majority of patients who are immunocompromised. Individuals with chronic renal disease, lymphoid malignancy, on B-cell targeted therapies, or with no serological response after two vaccine doses are at higher risk of poor response to a third vaccine dose. FUNDING Medical Research Council, Blood Cancer UK.
Collapse
Affiliation(s)
- Carl S Goodyear
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Amit Patel
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Eleanor Barnes
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Stefan Siebert
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Thushan I de Silva
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, The University of Sheffield, Sheffield, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Sean H Lim
- Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Sarah J Bowden
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Lucinda Billingham
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Alex Richter
- Clinical Immunology Service, University of Birmingham, Edgbaston, Birmingham, UK
| | - Miles Carroll
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | - Daniel Rea
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Helen Parry
- Department of Haematology, University Hospitals Birmingham NHS Foundations Trust, Birmingham, UK
| | - Sarah Pirrie
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Zixiang Lim
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jack Satsangi
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Gordon Cook
- National Institute for Health Research Leeds MIC, University of Leeds, Leeds, UK
| | - Paul Miller
- British Society of Blood and Marrow Transplantation and Cellular Therapy, Guy's Hospital, London, UK
| | - Neil Basu
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Ashley Gilmour
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Anne-Marie Hodgkins
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Lili Evans
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Ana Hughes
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Stephanie Longet
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK; Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, CNRS, Lyon, France
| | - Georgina Meacham
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kwee L Yong
- Cancer Institute, Department of Haematology, University College London, London, UK
| | | | - Mickey B C Koh
- Infection and Immunity Clinical Academic Group, St George's, University of London and Department of Haematology, St George's Hospital NHS Foundation Trust, London, UK
| | - Siobhan O Burns
- Clinical Immunology, Royal Free Hospital, Hampstead, London, UK; Institute of Immunity and Transplantation, University College London, Hampstead, London, UK
| | - Kim Orchard
- Department of Haematology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Caron Paterson
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Graham McIlroy
- Department of Haematology, University Hospitals Birmingham NHS Foundations Trust, Birmingham, UK
| | - Sam M Murray
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tina Thomson
- Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | | | - Lyndsey Goulston
- National Institute of Health Research, Southampton Clinical Research Facility, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Samantha Miller
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Victoria Keillor
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - David Thomas
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Amanda Kirkham
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK
| | - Iain B McInnes
- College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Pamela Kearns
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Edgbaston, Birmingham, UK; National Institute for Health Research, Birmingham Biomedical Research Centre, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
5
|
Wagner A, Garner-Spitzer E, Auer C, Gattinger P, Zwazl I, Platzer R, Orola-Taus M, Pichler P, Amman F, Bergthaler A, Huppa JB, Stockinger H, Zielinski CC, Valenta R, Kundi M, Wiedermann U. Breakthrough Infections in SARS-CoV-2-Vaccinated Multiple Myeloma Patients Improve Cross-Protection against Omicron Variants. Vaccines (Basel) 2024; 12:518. [PMID: 38793769 PMCID: PMC11125692 DOI: 10.3390/vaccines12050518] [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: 03/27/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
Patients with multiple myeloma (MM) are a heterogenous, immunocompromised group with increased risk for COVID-19 morbidity and mortality but impaired responses to primary mRNA SARS-CoV-2 vaccination. The effects of booster vaccinations and breakthrough infections (BTIs) on antibody (Ab) levels and cross-protection to variants of concern (VOCs) are, however, not sufficiently evaluated. Therefore, we analysed humoral and cellular vaccine responses in MM patients stratified according to disease stage/treatment into group (1) monoclonal gammopathy of undetermined significance, (2) after stem cell transplant (SCT) without immunotherapy (IT), (3) after SCT with IT, and (4) progressed MM, and in healthy subjects (prospective cohort study). In contrast to SARS-CoV-2 hu-1-specific Ab levels, Omicron-specific Abs and their cross-neutralisation capacity remained low even after three booster doses in a majority of MM patients. In particular, progressed MM patients receiving anti-CD38 mAb and those after SCT with IT were Ab low responders and showed delayed formation of spike-specific B memory cells. However, MM patients with hybrid immunity (i.e., vaccination and breakthrough infection) had improved cross-neutralisation capacity against VOCs, yet in the absence of severe COVID-19 disease. Our results indicate that MM patients require frequent variant-adapted booster vaccinations and/or changes to other vaccine formulations/platforms, which might have similar immunological effects as BTIs.
Collapse
Affiliation(s)
- Angelika Wagner
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, 1090 Vienna, Austria; (A.W.); (E.G.-S.); (C.A.); (I.Z.); (M.O.-T.); (P.P.)
| | - Erika Garner-Spitzer
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, 1090 Vienna, Austria; (A.W.); (E.G.-S.); (C.A.); (I.Z.); (M.O.-T.); (P.P.)
| | - Claudia Auer
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, 1090 Vienna, Austria; (A.W.); (E.G.-S.); (C.A.); (I.Z.); (M.O.-T.); (P.P.)
| | - Pia Gattinger
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria; (P.G.); (R.V.)
| | - Ines Zwazl
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, 1090 Vienna, Austria; (A.W.); (E.G.-S.); (C.A.); (I.Z.); (M.O.-T.); (P.P.)
| | - René Platzer
- Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University Vienna, 1090 Vienna, Austria; (R.P.); (F.A.); (A.B.); (J.B.H.); (H.S.)
| | - Maria Orola-Taus
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, 1090 Vienna, Austria; (A.W.); (E.G.-S.); (C.A.); (I.Z.); (M.O.-T.); (P.P.)
| | - Peter Pichler
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, 1090 Vienna, Austria; (A.W.); (E.G.-S.); (C.A.); (I.Z.); (M.O.-T.); (P.P.)
| | - Fabian Amman
- Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University Vienna, 1090 Vienna, Austria; (R.P.); (F.A.); (A.B.); (J.B.H.); (H.S.)
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, 1090 Vienna, Austria
| | - Andreas Bergthaler
- Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University Vienna, 1090 Vienna, Austria; (R.P.); (F.A.); (A.B.); (J.B.H.); (H.S.)
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, 1090 Vienna, Austria
| | - Johannes B. Huppa
- Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University Vienna, 1090 Vienna, Austria; (R.P.); (F.A.); (A.B.); (J.B.H.); (H.S.)
| | - Hannes Stockinger
- Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Medical University Vienna, 1090 Vienna, Austria; (R.P.); (F.A.); (A.B.); (J.B.H.); (H.S.)
| | - Christoph C. Zielinski
- Wiener Privatklinik, and Central European Cooperative Oncology Group (CECOG), Central European Cancer Center, 1090 Vienna, Austria;
| | - Rudolf Valenta
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria; (P.G.); (R.V.)
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Michael Kundi
- Center for Public Health, Medical University Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, 1090 Vienna, Austria; (A.W.); (E.G.-S.); (C.A.); (I.Z.); (M.O.-T.); (P.P.)
| |
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
|
Sehgal ANA, Safran J, Kratzer B, Gattinger P, Stieger RB, Musiejovsky L, Trapin D, Ettel P, Körmöczi U, Rottal A, Borochova K, Dorofeeva Y, Tulaeva I, Weber M, Grabmeier-Pfistershammer K, Perkmann T, Wiedermann U, Valenta R, Pickl WF. Flow Cytometry-Based Measurement of Antibodies Specific for Cell Surface-Expressed Folded SARS-CoV-2 Receptor-Binding Domains. Vaccines (Basel) 2024; 12:377. [PMID: 38675759 PMCID: PMC11053794 DOI: 10.3390/vaccines12040377] [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: 03/07/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has now become endemic and is currently one of the important respiratory virus infections regularly affecting mankind. The assessment of immunity against SARS-CoV-2 and its variants is important for guiding active and passive immunization and SARS-CoV-2-specific treatment strategies. METHODS We here devised a novel flow cytometry-based diagnostic platform for the assessment of immunity against cell-bound virus antigens. This platform is based on a collection of HEK-293T cell lines which, as exemplified in our study, stably express the receptor-binding domains (RBDs) of the SARS-CoV-2 S-proteins of eight major SARS-CoV-2 variants, ranging from Wuhan-Hu-1 to Omicron. RESULTS RBD-expressing cell lines stably display comparable levels of RBD on the surface of HEK-293T cells, as shown with anti-FLAG-tag antibodies directed against a N-terminally introduced 3x-FLAG sequence while the functionality of RBD was proven by ACE2 binding. We exemplify the usefulness and specificity of the cell-based test by direct binding of IgG and IgA antibodies of SARS-CoV-2-exposed and/or vaccinated individuals in which the assay shows a wide linear performance range both at very low and very high serum antibody concentrations. In another application, i.e., antibody adsorption studies, the test proved to be a powerful tool for measuring the ratios of individual variant-specific antibodies. CONCLUSION We have established a toolbox for measuring SARS-CoV-2-specific immunity against cell-bound virus antigens, which may be considered as an important addition to the armamentarium of SARS-CoV-2-specific diagnostic tests, allowing flexible and quick adaptation to new variants of concern.
Collapse
Affiliation(s)
- Al Nasar Ahmed Sehgal
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Jera Safran
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Pia Gattinger
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Robert B. Stieger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Laszlo Musiejovsky
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Doris Trapin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Paul Ettel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Ulrike Körmöczi
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Arno Rottal
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Kristina Borochova
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Yulia Dorofeeva
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Inna Tulaeva
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Milena Weber
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Grabmeier-Pfistershammer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria (J.S.); (R.B.S.)
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| |
Collapse
|
8
|
Vijayan A, Sukumaran A, Jones S, Paul A, Ahmed S, Mehta P, Mohanan M, Kumar S, Easwaran S, Shenoy P. Boosting Vaccine Response in Autoimmune Rheumatic Disease Patients With Inadequate Seroconversion: An Analysis of the Immunogenicity of Vector-Based and Inactivated Vaccines. Cureus 2024; 16:e55764. [PMID: 38586774 PMCID: PMC10998979 DOI: 10.7759/cureus.55764] [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] [Accepted: 03/06/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND An additional dose of COVID-19 vaccine is being offered to vaccinated people, especially those immunocompromised. The most widely available vaccines in India are the adenoviral vector-based AZD1222 (ChAdOx1 nCoV-19) and the heat-inactivated (BBV152). This study investigated the efficacy of both vaccines in patients with autoimmune rheumatic diseases (AIRD). OBJECTIVES To compare final anti-SARS-CoV-2 antibody titers, neutralization of pseudovirions by these antibodies, and T cell responses between patients of AIRD who had received the third dose of AZD1222 and BBV152 vaccines. METHODS Patients with stable AIRD who had completed two doses of COVID-19 vaccination but had a suboptimal response (anti-receptor binding domain (RBD) antibody<212) were randomized (1:1) to receive either AZD1222 or BBV152 as a booster dose. Patients with previous hybrid immunity or those who developed COVID-19 during the trial were excluded. Antibody titers, neutralization of Wuhan and Omicron pseudovirions, and interferon release by T cells (enzyme-linked immunosorbent spot (ELISpot)) in response to the Spike antigen were measured four weeks after this booster dose. RESULTS 146 were screened, 91 were randomized, and 67 were analyzed per protocol. The third dose improved antibody titers (p<0.001), neutralization of the Wuhan strain (p<0.001), and T cell interferon release (p<0.001) but not neutralization of the Omicron strain (p=0.24). Antibody titers were higher (p<0.005) after ADZ1222 boost (2,414 IU (interquartile range (IQR): 330-10,315)) than BBV1222 (347.7 IU (0.4-973)). Neutralization of the Wuhan stain was better (AZD1222: 76.6%(23.0-95.45) versus BBV152 (32.7% (0-78.9), p=0.03 by ANCOVA). Neutralization of Omicron (0 (0-28.4) vs 0 (0-4.8)) and T cell interferon release (57.0 IU (23.5-95) vs 50.5 IU (13.2-139)) were similar. CONCLUSION The third dose improved all parameters of immunogenicity in AIRD patients with previous inadequate responses except Omicron neutralization. The vector-based vaccine exhibits notable efficacy, particularly in antibody titers and neutralizing the Wuhan strain. TRIAL REGISTRATION CTRI/2021/12/038928.
Collapse
Affiliation(s)
- Anuroopa Vijayan
- Rheumatology, Dr Shenoys CARE, Kochi, IND
- Rheumatology, Sree Sudheendra Medical Mission, Kochi, IND
| | | | - Sara Jones
- Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, IND
| | - Aby Paul
- Pharmacy, Dr Shenoys CARE, Kochi, IND
| | - Sakir Ahmed
- Rheumatology, Kalinga Institute of Medical Sciences, Bhubaneswar, IND
| | - Pankti Mehta
- Clinical Immunology and Rheumatology, King George's Medical University, Lucknow, IND
| | | | - Santhosh Kumar
- Cancer Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, IND
| | | | | |
Collapse
|
9
|
Chen Z, Wang Y, He T, Li H, Ao L, Pan Q, Zhou Y, Zhu Q, Xiang D, Zhang G, Ling N, Chen M, Hu P, Peng M, Cai D, Zhang D, Ren H. Safety and Immunogenicity After Primary and Booster Inactivated SARS-Cov-2 Vaccination in Patients with Autoimmune Liver Diseases. J Clin Transl Hepatol 2024; 12:162-171. [PMID: 38343613 PMCID: PMC10851071 DOI: 10.14218/jcth.2023.00049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 01/05/2025] Open
Abstract
BACKGROUND AND AIMS SARS-CoV-2 vaccines-associated autoimmune liver diseases have been reported in several case reports. However, the safety and immunogenicity after primary and booster inactivated SARS-CoV-2 vaccination in patients with autoimmune liver diseases (AILD) is still unknown. METHODS Eighty-four patients with AILD were prospectively followed up after the second dose (primary) of inactivated SARS-CoV-2 vaccine. Some of them received the third dose (booster) of inactivated vaccine. Adverse events (AEs), autoimmune activation, and liver inflammation exacerbation after primary and booster vaccination were recorded. Meanwhile, dynamics of antireceptor-binding-domain IgG (anti-RBD-IgG), neutralizing antibodies (NAbs) and RBD-specific B cells responses were evaluated. RESULTS The overall AEs in AILD patients after primary and booster vaccination were 26.2% and 13.3%, respectively. The decrease of C3 level and increase of immunoglobulin light chain κ and λ levels were observed in AILD patients after primary vaccination, however, liver inflammation was not exacerbated, even after booster vaccination. Both the seroprevalence and titers of anti-RBD-IgG and NAbs were decreased over time in AILD patients after primary vaccination. Notably, the antibody titers were significantly elevated after booster vaccination (10-fold in anti-RBD-IgG and 7.4-fold in NAbs, respectively), which was as high as in healthy controls. Unfortunately, the inferior antibody response was not enhanced after booster vaccination in patients with immunosuppressants. Changes of atypical memory B cells were inversely related to antibody levels, which indicate that the impaired immune memory was partially restored partly by the booster vaccination. CONCLUSIONS The well tolerability and enhanced humoral immune response of inactivated vaccine supports an additional booster vaccination in AILD patients without immunosuppressants.
Collapse
Affiliation(s)
- Zhiwei Chen
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuting Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Taiyu He
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hu Li
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Ao
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingbo Pan
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingzhi Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Zhu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dejuan Xiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gaoli Zhang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ning Ling
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingli Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dachuan Cai
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dazhi Zhang
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Ren
- Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Schell TL, Caldera F. A Practical Update on COVID-19 and Inflammatory Bowel Disease: COVID-19 Disease Risk and Vaccine Safety and Efficacy. Gastroenterol Hepatol (N Y) 2024; 20:88-97. [PMID: 38414911 PMCID: PMC10895915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The COVID-19 pandemic introduced significant uncertainty regarding the care of patients with inflammatory bowel disease (IBD). Substantial research efforts have made progress in answering many of the questions that arose, but the constantly shifting paradigm of COVID-19-related research and recommendations has made it challenging for IBD clinicians to remain up-to-date. The goal of this article is to provide a concise and practical summary of the literature evaluating COVID-19 disease risk in addition to COVID-19 vaccine safety, immunogenicity, real-world effectiveness, and uptake among patients with IBD.
Collapse
Affiliation(s)
- Trevor L. Schell
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Freddy Caldera
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
11
|
Rabdano S, Ruzanova E, Makarov D, Vertyachikh A, Teplykh V, Rudakov G, Pletyukhina I, Saveliev N, Zakharov K, Alpenidze D, Vasilyuk V, Arakelov S, Skvortsova V. Safety and Immunogenicity of the Convacell ® Recombinant N Protein COVID-19 Vaccine. Vaccines (Basel) 2024; 12:100. [PMID: 38276672 PMCID: PMC10821050 DOI: 10.3390/vaccines12010100] [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: 12/06/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
We have developed Convacell®-a COVID-19 vaccine based on the recombinant nucleocapsid (N) protein of SARS-CoV-2. This paper details Convacell's® combined phase I/II and IIb randomized, double-blind, interventional clinical trials. The primary endpoints were the frequency of adverse effects (AEs) and the titers of specific anti-N IgGs induced by the vaccination; secondary endpoints included the nature of the immune response. Convacell® demonstrated high safety in phase I with no severe AEs detected, 100% seroconversion by day 42 and high and sustained for 350 days anti-N IgG levels in phase II. Convacell® also demonstrated a fused cellular and humoral immune response. Phase IIb results showed significant post-vaccination increases in circulating anti-N IgG and N protein-specific IFNγ+-producing PBMC quantities among 438 volunteers. Convacell® showed same level of immunological efficacy for single and double dose vaccination regimens, including for elderly patients. The clinical studies indicate that Convacell® is safe and highly immunogenic.
Collapse
Affiliation(s)
- Sevastyan Rabdano
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | - Ellina Ruzanova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | - Denis Makarov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | - Anastasiya Vertyachikh
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | - Valeriya Teplykh
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | - German Rudakov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | - Iuliia Pletyukhina
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | - Nikita Saveliev
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | | | - Diana Alpenidze
- State Budgetary Health Institution “City Polyclinic No. 117”, St. Petersburg 194358, Russia
| | - Vasiliy Vasilyuk
- Department of Toxicology, Extreme and Diving Medicine, North-Western State Medical University named after I.I. Mechnikov, St. Petersburg 191015, Russia
| | - Sergei Arakelov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia (I.P.)
| | | |
Collapse
|
12
|
Doherty J, O'Morain N, Stack R, Tosetto M, Inzitiari R, O'Reilly S, Gu L, Sheridan J, Cullen G, Mc Dermott E, Buckley M, Horgan G, Mulcahy H, Walshe M, Ryan EJ, Gautier V, Prostko J, Frias E, Daghfal D, Doran P, O'Morain C, Doherty GA. Reduced Serological Response to COVID-19 Booster Vaccine is Associated with Reduced B Cell Memory in Patients With Inflammatory Bowel Disease; VARIATION [VAriability in Response in IBD AgainsT SARS-COV-2 ImmunisatiON]. J Crohns Colitis 2023; 17:1445-1456. [PMID: 37018462 DOI: 10.1093/ecco-jcc/jjad065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 04/07/2023]
Abstract
BACKGROUND AND AIMS Patients with inflammatory bowel disease [IBD] have an attenuated response to initial COVID-19 vaccination. We sought to characterize the impact of IBD and its treatment on responses after the third vaccine against SARS-CoV-2. METHODS This was a prospective multicentre observational study of patients with IBD [n = 202] and healthy controls [HC, n = 92]. Serological response to vaccination was assessed by quantification of anti-spike protein [SP] immunoglobulin [Ig]G levels [anti-SPIgG] and in vitro neutralization of binding to angiotensin-converting enzyme 2 [ACE2]. Peripheral blood B-cell phenotype populations were assessed by flow cytometry. SARS-CoV-2 antigen-specific B-cell responses were assessed in ex vivo culture. RESULTS Median anti-SP IgG post-third vaccination in our IBD cohort was significantly lower than HCs [7862 vs 19 622 AU/mL, p < 0.001] as was ACE2 binding inhibition [p < 0.001]. IBD patients previously infected with COVID-19 [30%] had similar quantitative antibody response as HCs previously infected with COVID-19 [p = 0.12]. Lowest anti-SP IgG titres and neutralization were seen in IBD patients on anti-tumour necrosis factor [anti-TNF] agents, without prior COVID-19 infection, but all IBD patients show an attenuated vaccine response compared to HCs. Patients with IBD have reduced memory B-cell populations and attenuated B-cell responses to SARS-CoV-2 antigens if not previously infected with COVID-19 [p = 0.01]. Higher anti-TNF drug levels and zinc levels <65 ng/ml were associated with significantly lower serological responses. CONCLUSIONS Patients with IBD have an attenuated response to three doses of SARS-CoV-2 vaccine. Physicians should consider patients with higher anti-TNF drug levels and/or zinc deficiency as potentially at higher risk of attenuated response to vaccination.
Collapse
Affiliation(s)
- Jayne Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Neil O'Morain
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Roisin Stack
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Miriam Tosetto
- School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Sophie O'Reilly
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - Lili Gu
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - Juliette Sheridan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Garret Cullen
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Edel Mc Dermott
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Maire Buckley
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- St Michaels Hospital, Dun Laoghaire, Co Dublin, Ireland
| | - Gareth Horgan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- St Columcille's Hospital, Loughlinstown, Co Dublin, Ireland
| | - Hugh Mulcahy
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Margaret Walshe
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Elizabeth J Ryan
- Department of Biological Sciences, Health Research Institute, University of Limerick, Limerick, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - John Prostko
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - Edwin Frias
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - David Daghfal
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - Peter Doran
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Colm O'Morain
- Beacon Hospital, Sandyford, Co. Dublin and Trinity College Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Glen A Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| |
Collapse
|
13
|
Garner-Spitzer E, Wagner A, Gudipati V, Schoetta AM, Orola-Taus M, Kundi M, Kunert R, Mayrhofer P, Huppa JB, Stockinger H, Carsetti R, Gattinger P, Valenta R, Kratzer B, Sehgal ANA, Pickl WF, Reinisch W, Novacek G, Wiedermann U. Lower magnitude and faster waning of antibody responses to SARS-CoV-2 vaccination in anti-TNF-α-treated IBD patients are linked to lack of activation and expansion of cTfh1 cells and impaired B memory cell formation. EBioMedicine 2023; 96:104788. [PMID: 37672867 PMCID: PMC10485594 DOI: 10.1016/j.ebiom.2023.104788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Patients with inflammatory bowel disease (IBD) and healthy controls received primary SARS-CoV-2-mRNA vaccination and a booster after six months. Anti-TNF-α-treated patients showed significantly lower antibody (Ab) levels and faster waning than α4β7-integrin-antagonist recipients and controls. This prospective cohort study aimed to elucidate the underlying mechanisms on the basis of circulating T-follicular helper cells (cTfh) and B memory cells. METHODS We measured SARS-CoV-2- Wuhan and Omicron specific Abs, B- and T-cell subsets at baseline and kinetics of Spike (S)-specific B memory cells along with distributions of activated cTfh subsets before and after primary and booster vaccination. FINDINGS Lower and faster waning of Ab levels in anti-TNF-α treated IBD patients was associated with low numbers of total and naïve B cells vs. expanded plasmablasts prior to vaccination. Along with their low Ab levels against Wuhan and Omicron VOCs, reduced S-specific B memory cells were identified after the 2nd dose which declined to non-detectable after 6 months. In contrast, IBD patients with α4β7-integrin-antagonists and controls mounted and retained high Ab levels after the 2nd dose, which was associated with a pronounced increase in S-specific B memory cells that were maintained or expanded up to 6 months. Booster vaccination led to a strong increase of Abs with neutralizing capacity and S-specific B memory cells in these groups, which was not the case in anti-TNF-α treated IBD patients. Of note, Ab levels and S-specific B memory cells in particular post-booster correlated with the activation of cTfh1 cells after primary vaccination. INTERPRETATIONS The reduced magnitude, persistence and neutralization capacity of SARS-CoV-2 specific Abs after vaccination in anti-TNF-α-treated IBD patients were associated with impaired formation and maintenance of S-specific B memory cells, likely due to absent cTfh1 activation leading to extra-follicular immune responses and diminished B memory cell diversification. These observations have implications for patient-tailored vaccination schedules/vaccines in anti-TNF-α-treated patients, irrespective of their underlying disease. FUNDING The study was funded by third party funding of the Institute of Specific Prophylaxis and Tropical Medicine at the Medical University Vienna. The funders had no role in study design, data collection, data analyses, interpretation, or writing of report.
Collapse
Affiliation(s)
- Erika Garner-Spitzer
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria.
| | - Angelika Wagner
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria
| | - Venugopal Gudipati
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Anna-Margarita Schoetta
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Maria Orola-Taus
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria
| | - Michael Kundi
- Medical University of Vienna, Center for Public Health, Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Patrick Mayrhofer
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Johannes B Huppa
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Hannes Stockinger
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Rita Carsetti
- B Cell Research Unit, Immunology Research Area, Bambino Gesu Children's Hospital, IRCCS; Rome, Italy
| | - Pia Gattinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Vienna, Austria
| | - Rudolf Valenta
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Vienna, Austria; Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Bernhard Kratzer
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Al Nasar Ahmed Sehgal
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Winfried F Pickl
- Karl Landsteiner University of Health Sciences, Krems, Austria; Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Walter Reinisch
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gottfried Novacek
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria.
| |
Collapse
|
14
|
Ivanov N, Krastev B, Miteva DG, Batselova H, Alexandrova R, Velikova T. Effectiveness and safety of COVID-19 vaccines in patients with oncological diseases: State-of-the-art. World J Clin Oncol 2023; 14:343-356. [PMID: 37771630 PMCID: PMC10523189 DOI: 10.5306/wjco.v14.i9.343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/06/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023] Open
Abstract
Although the coronavirus disease 2019 (COVID-19) pandemic was declared to be no longer “a public health emergency of international concern” with its wide range of clinical manifestations and late complications, severe acute respiratory syndrome coronavirus 2 infection proved to be a serious threat, especially to the elderly and patients with comorbidities. Patients with oncologic diseases are vulnerable to severe infection and death. Indeed, patients with oncohematological diseases have a higher risk of severe COVID-19 and impaired post-vaccination immunity. Unfortunately, cancer patients are usually excluded from vaccine trials and investigations of post-vaccinal immune responses and the effectiveness of the vaccines. We aimed to elucidate to what extent patients with cancer are at increased risk of developing severe COVID-19 and what is their overall case fatality rate. We also present the current concept and evidence on the effectiveness and safety of COVID-19 vaccines, including boosters, in oncology patients. In conclusion, despite the considerably higher mortality in the cancer patient group than the general population, countries with high vaccination rates have demonstrated trends toward improved survival of cancer patients early and late in the pandemic.
Collapse
Affiliation(s)
- Nedelcho Ivanov
- Department of Clinical Immunology with Stem Cell Bank, University Hospital Alexanrovska, Sofia 1431, Bulgaria
| | - Boris Krastev
- Medical Center Nadezhda, Medical Center Nadezhda, Sofia 1407, Bulgaria
| | | | - Hristiana Batselova
- Department of Epidemiology and Disaster Medicine, Medical University, Plovdiv, University Hospital St. George, Plovdiv 6000, Bulgaria
| | - Radostina Alexandrova
- Department of Pathology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia 1000, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
15
|
Widhani A, Hasibuan AS, Rismawati R, Maria S, Koesnoe S, Hermanadi MI, Ophinni Y, Yamada C, Harimurti K, Sari ANL, Yunihastuti E, Djauzi S. Efficacy, Immunogenicity, and Safety of COVID-19 Vaccines in Patients with Autoimmune Diseases: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2023; 11:1456. [PMID: 37766132 PMCID: PMC10535431 DOI: 10.3390/vaccines11091456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Patients with autoimmune diseases are among the susceptible groups to COVID-19 infection because of the complexity of their conditions and the side effects of the immunosuppressive drugs used to treat them. They might show impaired immunogenicity to COVID-19 vaccines and have a higher risk of developing COVID-19. Using a systematic review and meta-analysis, this research sought to summarize the evidence on COVID-19 vaccine efficacy, immunogenicity, and safety in patients with autoimmune diseases following predefined eligibility criteria. Research articles were obtained from an initial search up to 26 September 2022 from PubMed, Embase, EBSCOhost, ProQuest, MedRxiv, bioRxiv, SSRN, EuroPMC, and the Cochrane Center of Randomized Controlled Trials (CCRCT). Of 76 eligible studies obtained, 29, 54, and 38 studies were included in systematic reviews of efficacy, immunogenicity, and safety, respectively, and 6, 18, and 4 studies were included in meta-analyses for efficacy, immunogenicity, and safety, respectively. From the meta-analyses, patients with autoimmune diseases showed more frequent breakthrough COVID-19 infections and lower total antibody (TAb) titers, IgG seroconversion, and neutralizing antibodies after inactivated COVID-19 vaccination compared with healthy controls. They also had more local and systemic adverse events after the first dose of inactivated vaccination compared with healthy controls. After COVID-19 mRNA vaccination, patients with autoimmune diseases had lower TAb titers and IgG seroconversion compared with healthy controls.
Collapse
Affiliation(s)
- Alvina Widhani
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
- Department of Internal Medicine, Universitas Indonesia Hospital, Depok 16424, Indonesia
| | - Anshari Saifuddin Hasibuan
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| | - Retia Rismawati
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| | - Suzy Maria
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| | - Sukamto Koesnoe
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| | - Muhammad Ikrar Hermanadi
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| | - Youdiil Ophinni
- Division of Clinical Virology, Center for Infectious Diseases, Graduate School of Medicine, Kobe University, Kobe 650-0017, Japan;
- Department of Host Defense, Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
- Center for Southeast Asian Studies, Kyoto University, Kyoto 606-8304, Japan;
| | - Chika Yamada
- Center for Southeast Asian Studies, Kyoto University, Kyoto 606-8304, Japan;
| | - Kuntjoro Harimurti
- Geriatric Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia;
| | - Aldean Nadhyia Laela Sari
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| | - Evy Yunihastuti
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| | - Samsuridjal Djauzi
- Allergy and Clinical Immunology Division, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta 10430, Indonesia; (A.S.H.); (R.R.); (S.M.); (S.K.); (M.I.H.); (A.N.L.S.); (E.Y.); (S.D.)
| |
Collapse
|
16
|
Overheu O, Lendowski S, Quast DR, Kühn D, Vidal Blanco E, Kraeft AL, Steinmann E, Kourti E, Lugnier C, Steinmann J, Reinacher-Schick A, Pfaender S. Longitudinal data on humoral response and neutralizing antibodies against SARS-CoV-2 Omicron BA.1 and subvariants BA.4/5 and BQ.1.1 after COVID-19 vaccination in cancer patients. J Cancer Res Clin Oncol 2023; 149:10633-10644. [PMID: 37300723 PMCID: PMC10257184 DOI: 10.1007/s00432-023-04961-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
PURPOSE The SARS-CoV-2 Omicron variant of concern (VOC) and subvariants like BQ.1.1 demonstrate immune evasive potential. Little is known about the efficacy of booster vaccinations regarding this VOC and subvariants in cancer patients. This study is among the first to provide data on neutralizing antibodies (nAb) against BQ.1.1. METHODS Cancer patients at our center were prospectively enrolled between 01/2021 and 02/2022. Medical data and blood samples were collected at enrollment and before and after every SARS-CoV-2 vaccination, at 3 and 6 months. RESULTS We analyzed 408 samples from 148 patients (41% female), mainly with solid tumors (85%) on active therapy (92%; 80% chemotherapy). SARS-CoV-2 IgG and nAb titers decreased over time, however, significantly increased following third vaccination (p < 0.0001). NAb (ND50) against Omicron BA.1 was minimal prior and increased significantly after the third vaccination (p < 0.0001). ND50 titers against BQ.1.1 after the third vaccination were significantly lower than against BA.1 and BA.4/5 (p < 0.0001) and undetectable in half of the patients (48%). Factors associated with impaired immune response were hematologic malignancies, B cell depleting therapy and higher age. Choice of vaccine, sex and treatment with chemo-/immunotherapy did not influence antibody response. Patients with breakthrough infections had significantly lower nAb titers after both 6 months (p < 0.001) and the third vaccination (p = 0.018). CONCLUSION We present the first data on nAb against BQ.1.1 following the third vaccination in cancer patients. Our results highlight the threat that new emerging SARS-CoV-2 variants pose to cancer patients and support efforts to apply repeated vaccines. Since a considerable number of patients did not display an adequate immune response, continuing to exhibit caution remains reasonable.
Collapse
Affiliation(s)
- Oliver Overheu
- Department of Hematology and Oncology with Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany.
| | - Simon Lendowski
- Department of Hematology and Oncology with Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Daniel R Quast
- Department of Internal Medicine, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Daniel Kühn
- Department of Molecular and Medical Virology, Ruhr University, Bochum, Germany
| | - Elena Vidal Blanco
- Department of Molecular and Medical Virology, Ruhr University, Bochum, Germany
| | - Anna-Lena Kraeft
- Department of Hematology and Oncology with Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Eike Steinmann
- Department of Molecular and Medical Virology, Ruhr University, Bochum, Germany
| | - Eleni Kourti
- Department of Hematology and Oncology with Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Celine Lugnier
- Department of Hematology and Oncology with Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Joerg Steinmann
- Institute of Clinical Hygiene, Medical Microbiology and Infectiology, Klinikum Nürnberg, Paracelsus Medical University, Nuremberg, Germany
| | - Anke Reinacher-Schick
- Department of Hematology and Oncology with Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr University, Bochum, Germany
| |
Collapse
|
17
|
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: 1.5] [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
|
18
|
Ralise AEG, Camargo TM, Marson FAL. Phase 4 clinical trials in the era of the Coronavirus Disease (COVID-19) pandemic and their importance to optimize the COVID-19 vaccination. Hum Vaccin Immunother 2023; 19:2234784. [PMID: 37449956 DOI: 10.1080/21645515.2023.2234784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023] Open
Abstract
Since the appearance of SARS-CoV-2, the scientific community has worked relentlessly to gather enough information about the illness caused by this virus infection. Such great effort has resulted in increased scientific publication, including phase 4 clinical trials addressing the applicability of COVID-19 vaccines. In those trials that investigated the properties of the vaccine among participants with morbidities, mainly immunocompromised individuals, the safety was recommended, but in the presence of immunogenicity, such protection was considered of short and medium terms. It was also observed that a physically active lifestyle might increase the immunogenicity of the COVID-19 vaccination in patients with autoimmune rheumatic diseases and in immunocompromised patients. The coadministration of different types of vaccine such as the combination of the recombinant adenovirus type 5 (AD5)-vectored Convidecia as heterologous reinforcement vs. CoronaVac with homologous reinforcement in adults previously vaccinated with CoronaVac, as well as the coadministration of inactivated COVID-19 vaccine followed by the administration of the tetravalent influenza vaccine (Fragmented, Inactivated) and the pneumococcal vaccine 23 presented satisfactory immunogenicity. However, the heterologous reinforcement had better immunogenicity when compared to the homologous reinforcement. Simultaneous COVID-19 vaccination and vaccines against seasonal influenza did not raise safety issues, producing acceptable levels of adverse reactions and preserving the antibody responses against SARS-CoV-2. In the lot-to-lot consistency evaluation, CoronaVac was seen to induce an immune response considered relatively high, and the lots presented a similar profile of stability and immunogenicity, thus enabling their large-scale distribution. In brief, this article addressed, mainly, the importance of evaluating the immunological response in the COVID-19 vaccination in patients with specific health conditions (e.g., immunocompromised individuals) aiming at enabling adjustments to the vaccine calendar in national vaccination programs.
Collapse
Affiliation(s)
- Ana Eliza Garcia Ralise
- Laboratory of Molecular Biology and Genetics, São Francisco University, Bragança Paulista, Brazil
| | - Tais Mendes Camargo
- Laboratory of Molecular Biology and Genetics, São Francisco University, Bragança Paulista, Brazil
| | | |
Collapse
|
19
|
Silzle T, Kahlert CR, Albrich WC, Nigg S, Demmer Steingruber R, Driessen C, Fischer S. Humoral and cellular BNT162b2 mRNA-based booster vaccine-induced immunity in patients with multiple myeloma and persistence of neutralising antibodies: results of a prospective single-centre cohort study. Swiss Med Wkly 2023; 153:40090. [PMID: 37410944 DOI: 10.57187/smw.2023.40090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Currently available messenger ribonucleic acid (mRNA)-based vaccines against coronavirus disease (COVID-19) have been shown to be effective even in highly immunocompromised hosts, including patients with multiple myeloma. However, vaccination failure can be observed in all patient groups. METHODS This prospective study longitudinally assessed the humoral and cellular responses to a third booster dose of BNT162b2 mRNA-based vaccine in patients with myeloma (n = 59) and healthy controls (n = 22) by measuring the levels of anti-spike (S) antibodies (electro-chemiluminescence immunoassay) including neutralising antibodies and specific T-cells (enzyme-linked immunospot assay) following booster administration. RESULTS The third booster dose showed a high immunogenicity on the serological level among the patients with multiple myeloma (median anti-S level = 41 binding antibody units [BAUs]/ml pre-booster vs 3902 BAU/ml post-booster, p <0.001; increase in the median neutralising antibody level from 19.8% to 97%, p <0.0001). Four of five (80%) patients with a complete lack of any serological response (anti-S immunoglobulin level <0.8 BAU/ml) after two vaccine doses developed detectable anti-S antibodies after booster vaccination (median anti-S level = 88 BAU/ml post-booster). T-cell responses were largely preserved among the patients with multiple myeloma with no difference from the healthy controls following baseline vaccination (median spot-forming units [SFU]/106 of peripheral blood mononuclear cells = 193 vs 175, p = 0.711); these responses were augmented significantly after booster administration among the patients with multiple myeloma (median SFU/106 of peripheral blood mononuclear cells = 235 vs 443, p <0.001). However, the vaccination responses remained highly heterogeneous and diminished over time, with insufficient serological responses occurring even after booster vaccination in a few patients irrespective of the treatment intensity. CONCLUSIONS Our data demonstrate improvements in humoral and cellular immunity following booster vaccination and support the assessment of the humoral vaccine response in patients with multiple myeloma until a threshold for protection against severe COVID-19 is validated. This strategy can allow the identification of patients who might benefit from additional protective measures (e.g. pre-exposure prophylaxis via passive immunisation).
Collapse
Affiliation(s)
- Tobias Silzle
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Christian R Kahlert
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
- Infectious Diseases and Hospital Epidemiology, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Werner C Albrich
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Susanne Nigg
- Division of Infectious Diseases and Hospital Epidemiology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Ruth Demmer Steingruber
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Christoph Driessen
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Stefanie Fischer
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
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: 10.0] [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
|
Rabdano SO, Ruzanova EA, Pletyukhina IV, Saveliev NS, Kryshen KL, Katelnikova AE, Beltyukov PP, Fakhretdinova LN, Safi AS, Rudakov GO, Arakelov SA, Andreev IV, Kofiadi IA, Khaitov MR, Valenta R, Kryuchko DS, Berzin IA, Belozerova NS, Evtushenko AE, Truhin VP, Skvortsova VI. Immunogenicity and In Vivo Protective Effects of Recombinant Nucleocapsid-Based SARS-CoV-2 Vaccine Convacell ®. Vaccines (Basel) 2023; 11:vaccines11040874. [PMID: 37112786 PMCID: PMC10141225 DOI: 10.3390/vaccines11040874] [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: 04/02/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
The vast majority of SARS-CoV-2 vaccines which are licensed or under development focus on the spike (S) protein and its receptor binding domain (RBD). However, the S protein shows considerable sequence variations among variants of concern. The aim of this study was to develop and characterize a SARS-CoV-2 vaccine targeting the highly conserved nucleocapsid (N) protein. Recombinant N protein was expressed in Escherichia coli, purified to homogeneity by chromatography and characterized by SDS-PAGE, immunoblotting, mass spectrometry, dynamic light scattering and differential scanning calorimetry. The vaccine, formulated as a squalane-based emulsion, was used to immunize Balb/c mice and NOD SCID gamma (NSG) mice engrafted with human PBMCs, rabbits and marmoset monkeys. Safety and immunogenicity of the vaccine was assessed via ELISA, cytokine titer assays and CFSE dilution assays. The protective effect of the vaccine was studied in SARS-CoV-2-infected Syrian hamsters. Immunization induced sustainable N-specific IgG responses and an N-specific mixed Th1/Th2 cytokine response. In marmoset monkeys, an N-specific CD4+/CD8+ T cell response was observed. Vaccinated Syrian hamsters showed reduced lung histopathology, lower virus proliferation, lower lung weight relative to the body, and faster body weight recovery. Convacell® thus is shown to be effective and may augment the existing armamentarium of vaccines against COVID-19.
Collapse
Affiliation(s)
- Sevastyan O Rabdano
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Ellina A Ruzanova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Iuliia V Pletyukhina
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Nikita S Saveliev
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | | | | | - Petr P Beltyukov
- Scientific Research Institute of Hygiene, Occupational Pathology and Human Ecology of the Federal Medical-Biological Agency of Russia (SRIHOPHE), Kuzmolovsky 188663, Russia
| | - Liliya N Fakhretdinova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Ariana S Safi
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - German O Rudakov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Sergei A Arakelov
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Igor V Andreev
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
| | - Ilya A Kofiadi
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Immunology, N.I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow 117997, Russia
| | - Musa R Khaitov
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Immunology, N.I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow 117997, Russia
| | - Rudolf Valenta
- National Research Center Institute of Immunology (NRCII), Federal Medical-Biological Agency of Russia, Moscow 115522, Russia
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, Moscow 119435, Russia
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Daria S Kryuchko
- Federal Medical-Biological Agency of Russia, Moscow 125310, Russia
| | - Igor A Berzin
- Federal Medical-Biological Agency of Russia, Moscow 125310, Russia
| | - Natalia S Belozerova
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Anatoly E Evtushenko
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | - Viktor P Truhin
- Saint Petersburg Scientific Research Institute of Vaccines and Serums of the Federal Medical-Biological Agency of Russia (SPbSRIVS), St. Petersburg 198320, Russia
| | | |
Collapse
|
22
|
Liatsou E, Ntanasis-Stathopoulos I, Lykos S, Ntanasis-Stathopoulos A, Gavriatopoulou M, Psaltopoulou T, Sergentanis TN, Terpos E. Adult Patients with Cancer Have Impaired Humoral Responses to Complete and Booster COVID-19 Vaccination, Especially Those with Hematologic Cancer on Active Treatment: A Systematic Review and Meta-Analysis. Cancers (Basel) 2023; 15:cancers15082266. [PMID: 37190194 DOI: 10.3390/cancers15082266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
The exclusion of patients with cancer in clinical trials evaluating COVID-19 vaccine efficacy and safety, in combination with the high rate of severe infections, highlights the need for optimizing vaccination strategies. The aim of this study was to perform a systematic review and meta-analysis of the published available data from prospective and retrospective cohort studies that included patients with either solid or hematological malignancies according to the PRISMA Guidelines. A literature search was performed in the following databases: Medline (Pubmed), Scopus, Clinicaltrials.gov, EMBASE, CENTRAL and Google Scholar. Overall, 70 studies were included for the first and second vaccine dose and 60 studies for the third dose. The Effect Size (ES) of the seroconversion rate after the first dose was 0.41 (95%CI: 0.33-0.50) for hematological malignancies and 0.56 (95%CI: 0.47-0.64) for solid tumors. The seroconversion rates after the second dose were 0.62 (95%CI: 0.57-0.67) for hematological malignancies and 0.88 (95%CI: 0.82-0.93) for solid tumors. After the third dose, the ES for seroconversion was estimated at 0.63 (95%CI: 0.54-0.72) for hematological cancer and 0.88 (95%CI: 0.75-0.97) for solid tumors. A subgroup analysis was performed to evaluate potential factors affecting immune response. Production of anti-SARS-CoV-2 antibodies was found to be more affected in patients with hematological malignancies, which was attributed to the type of malignancy and treatment with monoclonal antibodies according to the subgroup analyses. Overall, this study highlights that patients with cancer present suboptimal humoral responses after COVID-19 vaccination. Several factors including timing of vaccination in relevance with active therapy, type of therapy, and type of cancer should be considered throughout the immunization process.
Collapse
Affiliation(s)
- Efstathia Liatsou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | | | - Stavros Lykos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | | | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Theodora Psaltopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Theodoros N Sergentanis
- Department of Public Health Policy, School of Public Health, University of West Attica, 12243 Aigaleo, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
23
|
Purshouse K, Thomson JP, Vallet M, Alexander L, Bonisteel I, Brennan M, Cameron DA, Figueroa JD, Furrie E, Haig P, Heck M, McCaughan H, Mitchell P, McVicars H, Primrose L, Silva I, Templeton K, Wilson N, Hall PS. The Scottish COVID Cancer Immunity Prevalence Study: A Longitudinal Study of SARS-CoV-2 Immune Response in Patients Receiving Anti-Cancer Treatment. Oncologist 2023; 28:e145-e155. [PMID: 36719033 PMCID: PMC10020811 DOI: 10.1093/oncolo/oyac257] [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: 07/01/2022] [Accepted: 10/27/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Cancer and anti-cancer treatment (ACT) may be risk factors for severe SARS-CoV-2 infection and limited vaccine efficacy. Long-term longitudinal studies are needed to evaluate these risks. The Scottish COVID cancer immunity prevalence (SCCAMP) study characterizes the incidence and outcomes of SARS-CoV-2 infection and vaccination in patients with solid tumors undergoing ACT. This preliminary analysis includes 766 patients recruited since May 2020. METHODS Patients with solid-organ cancers attending secondary care for active ACT consented to the collection of routine electronic health record data and serial blood samples over 12 months. Blood samples were tested for total SARS-CoV-2 antibody. RESULTS A total of 766 participants were recruited between May 28, 2020 and October 31, 2021. Most received cytotoxic chemotherapy (79%). Among the participants, 48 (6.3%) were tested positive for SARS-CoV-2 by PCR. Infection rates were unaffected by ACT, largely aligning with the local population. Mortality proportion was not higher with a recent positive SARS-CoV-2 PCR (10.4% vs 10.6%). Multivariate analysis revealed lower infection rates in vaccinated patients regardless of chemotherapy (HR 0.307 [95% CI, 0.144-0.6548]) or immunotherapy (HR 0.314 [95% CI, 0.041-2.367]) treatment. A total of 96.3% of patients successfully raised SARS-CoV-2 antibodies after >2 vaccines. This was independent of the treatment type. CONCLUSION This is the largest on-going longitudinal real-world dataset of patients undergoing ACT during the early stages of the COVID-19 pandemic. This preliminary analysis demonstrates that patients with solid tumors undergoing ACT have high protection from SARS-CoV-2 infection following COVID-19 vaccination. The SCCAMP study will evaluate long-term COVID-19 antibody trends, focusing on specific ACTs and patient subgroups.
Collapse
Affiliation(s)
| | | | | | - Lorna Alexander
- Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, UK
| | - Isaac Bonisteel
- The University of Edinburgh Medical School, The University of Edinburgh, Chancellor’s Building, Edinburgh BioQuarter, Edinburgh, UK
| | - Maree Brennan
- Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, UK
| | - David A Cameron
- Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, UK
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Jonine D Figueroa
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Edinburgh, UK
- Usher Institute, Centre for Population Health Sciences, Old Medical School, Teviot Place, Edinburgh, UK
| | - Elizabeth Furrie
- Department of Immunology, Ninewells Hospital and Dundee Medical School, Dundee, UK
| | - Pamela Haig
- Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, UK
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Mattea Heck
- The University of Edinburgh Medical School, The University of Edinburgh, Chancellor’s Building, Edinburgh BioQuarter, Edinburgh, UK
| | - Hugh McCaughan
- Clinical Infection Research Group, Regional Infectious Diseases Unit, Western General Hospital, Edinburgh, UK
| | - Paul Mitchell
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Heather McVicars
- Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, UK
| | | | - Ines Silva
- Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, UK
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Kate Templeton
- Clinical Infection Research Group, Regional Infectious Diseases Unit, Western General Hospital, Edinburgh, UK
| | - Natalie Wilson
- Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, UK
- Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Peter S Hall
- Corresponding author: Peter S. Hall, PhD, Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Edinburgh Cancer Centre, NHS Lothian, Crewe Road South, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
24
|
Gattinger P, Ohradanova-Repic A, Valenta R. Importance, Applications and Features of Assays Measuring SARS-CoV-2 Neutralizing Antibodies. Int J Mol Sci 2023; 24:ijms24065352. [PMID: 36982424 PMCID: PMC10048970 DOI: 10.3390/ijms24065352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/17/2023] Open
Abstract
More than three years ago, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) caused the unforeseen COVID-19 pandemic with millions of deaths. In the meantime, SARS-CoV-2 has become endemic and is now part of the repertoire of viruses causing seasonal severe respiratory infections. Due to several factors, among them the development of SARS-CoV-2 immunity through natural infection, vaccination and the current dominance of seemingly less pathogenic strains belonging to the omicron lineage, the COVID-19 situation has stabilized. However, several challenges remain and the possible new occurrence of highly pathogenic variants remains a threat. Here we review the development, features and importance of assays measuring SARS-CoV-2 neutralizing antibodies (NAbs). In particular we focus on in vitro infection assays and molecular interaction assays studying the binding of the receptor binding domain (RBD) with its cognate cellular receptor ACE2. These assays, but not the measurement of SARS-CoV-2-specific antibodies per se, can inform us of whether antibodies produced by convalescent or vaccinated subjects may protect against the infection and thus have the potential to predict the risk of becoming newly infected. This information is extremely important given the fact that a considerable number of subjects, in particular vulnerable persons, respond poorly to the vaccination with the production of neutralizing antibodies. Furthermore, these assays allow to determine and evaluate the virus-neutralizing capacity of antibodies induced by vaccines and administration of plasma-, immunoglobulin preparations, monoclonal antibodies, ACE2 variants or synthetic compounds to be used for therapy of COVID-19 and assist in the preclinical evaluation of vaccines. Both types of assays can be relatively quickly adapted to newly emerging virus variants to inform us about the magnitude of cross-neutralization, which may even allow us to estimate the risk of becoming infected by newly appearing virus variants. Given the paramount importance of the infection and interaction assays we discuss their specific features, possible advantages and disadvantages, technical aspects and not yet fully resolved issues, such as cut-off levels predicting the degree of in vivo protection.
Collapse
Affiliation(s)
- Pia Gattinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Karl Landsteiner University, 3500 Krems an der Donau, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
- NRC Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Correspondence:
| |
Collapse
|
25
|
Kaufman HW, Meyer WA, Clarke NJ, Radcliff J, Rank CM, Freeman J, Eisenberg M, Gillim L, Morice WG, Briscoe DM, Perlin DS, Wohlgemuth JG. Assessing Vulnerability to COVID-19 in High-Risk Populations: The Role of SARS-CoV-2 Spike-Targeted Serology. Popul Health Manag 2023; 26:29-36. [PMID: 36799932 DOI: 10.1089/pop.2022.0241] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Individuals at increased risk for severe coronavirus disease-2019 (COVID-19) outcomes, due to compromised immunity or other risk factors, would benefit from objective measures of vulnerability to infection based on vaccination or prior infection. The authors reviewed published data to identify a specific role and interpretation of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) spike-targeted serology testing. Specific recommendations are provided for an evidence-based and clinically-useful interpretation of SARS-CoV-2 spike-targeted serology to identify vulnerability to infection and potential subsequent adverse outcomes. Decreased vaccine effectiveness among immunocompromised individuals is linked to correspondingly high rates of breakthrough infections. Negative results on SARS-CoV-2 antibody tests are associated with increased risk for subsequent infection. "Low-positive" results on semiquantitative SARS-CoV-2 spike-targeted antibody tests may help identify persons at increased risk as well. Standardized SARS-CoV-2 spike-targeted antibody tests may provide objective information on the risk of SARS-CoV-2 infection and associated adverse outcomes. This holds especially for high-risk populations that demonstrate a relatively high rate of seronegativity. The widespread availability of such tests presents an opportunity to refine risk assessment for individuals with suboptimal SARS-CoV-2 antibody levels and to promote effective interventions. Interim federal guidance would support physicians and patients while additional investigations are pursued.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - David S Perlin
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey, USA
| | | |
Collapse
|
26
|
Woelfel S, Dütschler J, König M, Graf N, Oikonomou V, Krieger C, Truniger S, Franke A, Eckhold A, Forsch K, Wyss J, Krupka N, Albrich W, Frei N, Geissler N, Schaub P, Friedrich M, Misselwitz B, Korte W, Bürgi JJ, Brand S. Systemic and T cell-associated responses to SARS-CoV-2 immunisation in gut inflammation (STAR SIGN study): effects of biologics on vaccination efficacy of the third dose of mRNA vaccines against SARS-CoV-2. Aliment Pharmacol Ther 2023; 57:103-116. [PMID: 36307899 PMCID: PMC9874447 DOI: 10.1111/apt.17264] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 08/29/2022] [Accepted: 10/10/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Immunosuppressed patients with inflammatory bowel disease (IBD) experience increased risk of vaccine-preventable diseases such as COVID-19. AIMS To assess humoral and cellular immune responses following SARS-CoV-2 booster vaccination in immunosuppressed IBD patients and healthy controls. METHODS In this prospective, multicentre, case-control study, 139 IBD patients treated with biologics and 110 healthy controls were recruited. Serum anti-SARS-CoV-2 spike IgG concentrations were measured 2-16 weeks after receiving a third mRNA vaccine dose. The primary outcome was to determine if humoral immune responses towards booster vaccines differ in IBD patients under anti-TNF versus non-anti-TNF therapy and healthy controls. Secondary outcomes were antibody decline, impact of previous infection and SARS-CoV-2-targeted T cell responses. RESULTS Anti-TNF-treated IBD patients showed reduced anti-spike IgG concentrations (geometric mean 2357.4 BAU/ml [geometric SD 3.3]) when compared to non-anti-TNF-treated patients (5935.7 BAU/ml [3.9]; p < 0.0001) and healthy controls (5481.7 BAU/ml [2.4]; p < 0.0001), respectively. In multivariable modelling, prior infection (geometric mean ratio 2.00 [95% CI 1.34-2.90]) and vaccination with mRNA-1273 (1.53 [1.01-2.27]) increased antibody concentrations, while anti-TNF treatment (0.39 [0.28-0.54]) and prolonged time between vaccination and antibody measurement (0.72 [0.58-0.90]) decreased anti-SARS-CoV-2 spike antibodies. Antibody decline was comparable in IBD patients independent of anti-TNF treatment and antibody concentrations could not predict breakthrough infections. Cellular and humoral immune responses were uncoupled, and more anti-TNF-treated patients than healthy controls developed inadequate T cell responses (15/73 [20.5%] vs 2/100 [2.0%]; p = 0.00031). CONCLUSIONS Anti-TNF-treated IBD patients have impaired humoral and cellular immunogenicity following SARS-CoV-2 booster vaccination. Fourth dose administration may be beneficial for these patients.
Collapse
Affiliation(s)
- Simon Woelfel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of MedicineLudwig Maximilian University of MunichMunichGermany,Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Joel Dütschler
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland,Outpatient ClinicAmbulatory Services RorschachRorschachSwitzerland
| | - Marius König
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Nicole Graf
- Clinical Trials UnitCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Vasileios Oikonomou
- Department of Visceral Surgery and Medicine, Inselspital Bern University HospitalUniversity of BernBernSwitzerland
| | - Claudia Krieger
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Samuel Truniger
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland,Outpatient ClinicAmbulatory Services RorschachRorschachSwitzerland
| | - Annett Franke
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland,Outpatient ClinicAmbulatory Services RorschachRorschachSwitzerland
| | - Annika Eckhold
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Kristina Forsch
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Jacqueline Wyss
- Department of Visceral Surgery and Medicine, Inselspital Bern University HospitalUniversity of BernBernSwitzerland
| | - Niklas Krupka
- Department of Visceral Surgery and Medicine, Inselspital Bern University HospitalUniversity of BernBernSwitzerland
| | - Werner Albrich
- Department of Infectious DiseasesCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Nicola Frei
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Nora Geissler
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | - Peter Schaub
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| | | | - Matthias Friedrich
- Translational Gastroenterology Unit, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Benjamin Misselwitz
- Department of Visceral Surgery and Medicine, Inselspital Bern University HospitalUniversity of BernBernSwitzerland
| | | | | | - Stephan Brand
- Department of Gastroenterology and HepatologyCantonal Hospital St. GallenSt. GallenSwitzerland
| |
Collapse
|
27
|
Huf WE, Valipour A. Vaccination against SARS-CoV-2 in adults with a diagnosis of cancer: a short review. MEMO 2023; 16:73-78. [PMID: 36628356 PMCID: PMC9815670 DOI: 10.1007/s12254-022-00858-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/22/2022] [Indexed: 01/07/2023]
Abstract
Compared to individuals without cancer, patients with a diagnosis of malignancy bear a higher risk of becoming infected with SARS-CoV‑2, suffer more frequently from disease-related complications, and are more likely to die due to coronavirus disease 2019 (COVID-19). Depending on the type of cancer and the treatment received, the immune response to vaccination may also be affected in patients with certain types of malignancy. Therefore, there is a need for more specific COVID-19 vaccination recommendations in individuals with a diagnosis of cancer. Furthermore, pre-exposition prophylaxis should be considered for some patients. This short review summarizes some challenges in prevention of (severe) COVID-19 in individuals with a diagnosis of cancer and compares guidelines given by the US National Comprehensive Cancer Network, German Robert Koch-Institut, and Austrian Nationales Impfgremium.
Collapse
Affiliation(s)
- Waltraud Elisabeth Huf
- Department of Respiratory and Critical Care Medicine, Klinik Floridsdorf, Vienna Healthcare Group, Bruennerstr. 68, 1210 Vienna, Austria ,grid.487248.50000 0004 9340 1179Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna Healthcare Group, Bruennerstr. 68, 1210 Vienna, Austria
| | - Arschang Valipour
- Department of Respiratory and Critical Care Medicine, Klinik Floridsdorf, Vienna Healthcare Group, Bruennerstr. 68, 1210 Vienna, Austria ,grid.487248.50000 0004 9340 1179Karl-Landsteiner-Institute for Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna Healthcare Group, Bruennerstr. 68, 1210 Vienna, Austria
| |
Collapse
|
28
|
Full seroconversion in initial non-responders with higher antibody levels after heterologous COVID-19 vaccination schedule. Immunol Lett 2022; 250:1-6. [PMID: 36108774 PMCID: PMC9468054 DOI: 10.1016/j.imlet.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/23/2022] [Accepted: 09/11/2022] [Indexed: 11/21/2022]
Abstract
Antibody testing after COVID-19 vaccination is generally not recommended. Here, we present the results of a retrospective study, in which we analyzed antibody levels before and after the first dose of the ChAdOx1 vector vaccine. We identified 5% non-responders (43.6 ± 10.6 years; females: 41%) and 3.4% low-responders (44.2 ± 10.1 years; females: 64%) after the first dose. Of these, 61 individuals received a timely second dose either with a homologous (ChAdOx1/ChAdOx1) or heterologous (ChAdOx1/mRNA-1273) schedule. All vaccinees achieved positive S1-specific IgG titers to the ancestral SARS-CoV-2 strain after the second dose, but antibody levels as well as neutralization titers against the ancestral SARS-CoV-2 strain were higher after the heterologous schedule. However, Omicron-specific neutralizing antibodies were not detectable after two doses in either group, indicating that a third vaccine dose is needed to enhance cross-reactive antibodies against currently circulating and emerging variants of concern.
Collapse
|
29
|
Ohradanova-Repic A, Skrabana R, Gebetsberger L, Tajti G, Baráth P, Ondrovičová G, Praženicová R, Jantova N, Hrasnova P, Stockinger H, Leksa V. Blockade of TMPRSS2-mediated priming of SARS-CoV-2 by lactoferricin. Front Immunol 2022; 13:958581. [PMID: 36081512 PMCID: PMC9445877 DOI: 10.3389/fimmu.2022.958581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022] Open
Abstract
In addition to vaccines, there is an urgent need for supplemental antiviral therapeutics to dampen the persistent COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The transmembrane protease serine 2 (TMPRSS2), that is responsible for proteolytic priming of the SARS-CoV-2 spike protein, appears as a rational therapeutic target. Accordingly, selective inhibitors of TMPRSS2 represent potential tools for prevention and treatment of COVID-19. Previously, we identified the human milk glycoprotein lactoferrin as a natural inhibitor of plasminogen conversion to plasmin, a serine protease homologous to TMPRSS2. Here, we tested whether lactoferrin and lactoferricin, a biologically active natural peptide produced by pepsin-mediated digestion of lactoferrin, together with synthetic peptides derived from lactoferrin, were able to block TMPRSS2 and SARS-CoV-2 infection. Particularly, we revealed that both lactoferricin and the N-terminal synthetic peptide pLF1 significantly inhibited: i) proteolytic activity of TMPRSS2 and plasmin, ii) proteolytic processing of the SARS-CoV-2 spike protein, and iii) SARS-CoV-2 infection of SARS-CoV-2-permissive cells. Thus, natural and synthetic peptides derived from lactoferrin represent feasible candidates for supporting prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Anna Ohradanova-Repic
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
- *Correspondence: Vladimir Leksa, ; Anna Ohradanova-Repic,
| | - Rostislav Skrabana
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Laura Gebetsberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Gabor Tajti
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Peter Baráth
- Department of Glycobiology, Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Gabriela Ondrovičová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Romana Praženicová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Biochemistry, Comenius University, Bratislava, Slovakia
| | - Nikola Jantova
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Biochemistry, Comenius University, Bratislava, Slovakia
| | - Patricia Hrasnova
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Biochemistry, Comenius University, Bratislava, Slovakia
| | - Hannes Stockinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Vladimir Leksa
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
- *Correspondence: Vladimir Leksa, ; Anna Ohradanova-Repic,
| |
Collapse
|
30
|
Su E, Fischer S, Demmer-Steingruber R, Nigg S, Güsewell S, Albrich WC, Rothermundt C, Silzle T, Kahlert CR. Humoral and cellular responses to mRNA-based COVID-19 booster vaccinations in patients with solid neoplasms under active treatment. ESMO Open 2022; 7:100587. [PMID: 36156449 PMCID: PMC9399124 DOI: 10.1016/j.esmoop.2022.100587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 11/26/2022] Open
Abstract
Background Patients with cancer are at high risk for severe coronavirus disease 2019 (COVID-19) infection. Knowledge regarding the efficacy of the messenger RNA (mRNA) vaccines in actively treated cancer patients is limited as they had been excluded from the pivotal studies of these vaccines. We evaluated humoral and cellular immune responses in cancer patients after double vaccination and a booster dose and identified disease- and treatment-related factors associated with a reduced immune response. We also documented the number and outcome of breakthrough infections. Patients and methods Patients with metastatic solid malignancies undergoing active treatment were included if they had received two doses of the severe acute respiratory syndrome coronavirus 2 mRNA vaccines BNT162b2 or mRNA-1273 and a booster dose. Other causes of immunosuppression and previous COVID-19 infections (positive anti-nucleocapsid titers) were exclusion criteria. Anti-spike antibodies, neutralizing antibodies (nAbs) and T-cell responses were assessed about 6 months after the two-dose vaccination and 4 weeks after the booster. Results Fifty-one patients had pre-booster and 46 post-booster measurements. Anti-spike titers after two vaccine doses were highly variable and significantly lower in older patients, during treatment with chemotherapy compared to targeted and endocrine treatments and in patients with low CD4+ or CD19+ cell counts. The booster dose led to a significant increase in anti-spike antibodies and nAbs, achieving almost uniformly high titers, irrespective of baseline and treatment factors. The cellular immune response was also significantly increased by the booster, however generally more stable and not influenced by baseline factors and treatment type. Seventeen patients (33%) experienced breakthrough infections, but none required hospital care or died from COVID-19. Conclusions An mRNA vaccine booster dose is able to increase humoral and cellular immune responses and to overcome the immunosuppressive influence of baseline and treatment factors in cancer patients. Breakthrough infections were uniformly mild in this vaccinated high-risk population. We provide data on humoral and cellular immunity after SARS-CoV-2 mRNA vaccination in actively treated cancer patients. We found highly variable anti-S titers after two doses; titers were influenced by several baseline and treatment factors. The booster achieved uniformly higher anti-S titers; the influence of baseline factors could be overcome by the booster. Spike-specific T-cell immunity was more stable and not influenced by baseline factors. Even though many breakthrough infections were recorded during the Omicron wave, they were uniformly mild.
Collapse
|
31
|
Second Booster BNT162b2 Restores SARS-CoV-2 Humoral Response in Patients With Multiple Myeloma, Excluding Those Under Anti-BCMA Therapy. Hemasphere 2022; 6:e764. [PMID: 35928542 PMCID: PMC9345642 DOI: 10.1097/hs9.0000000000000764] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 07/08/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 vaccination leads to a less intense humoral response in patients with multiple myeloma (MM) compared with healthy individuals, whereas the SARS-CoV-2-specific immunity fades over time. The purpose of this study was to explore the kinetics of SARS-CoV-2 neutralizing antibodies (NAbs) in patients with MM after vaccination with the BNT162b2 mRNA vaccine, focusing on their response before (B4D) and at 1 month after the fourth vaccination (M1P4D). Overall, 201 patients with a median age of 67 years were included, whereas 114 (56.7%) were men. The median NAbs levels B4D were 80.0% (±3.5%) and at M1P4D they increased to a median value of 96.1% (±3.7%). The NAb values at M1P4D were similar to those at 1 month post the third dose and superior to all previous timepoints. At M1P4D, the NAbs levels of all the treatment groups increased, apart from the anti-BCMA group. A significant increase in median NAbs values was observed for those receiving CD38-based treatment (n = 43, from 71.0% B4D to 96.0% at M1P4D) and those who did not receive CD38- or BCMA-targeted therapy (n = 137, from 89.6% B4D to 96.3% at M1P4D). Regarding the patients under BCMA-based therapy (n = 21), there was no remarkable increase in NAbs values following the second booster shot (from 3.0% B4D to 4.0% at M1P4D). In conclusion, booster vaccination with the BNT162b2 results in a substantially improved humoral response against SARS-CoV-2 in patients with MM. Anti-BCMA treatment remains an adverse predictive factor for NAbs response; thus, tailored prevention measures should be considered for this patient subgroup.
Collapse
|