1
|
Rotundo S, Serapide F, Berardelli L, Gullì SP, Mongiardi S, Tassone MT, Trecarichi EM, Russo A. Early combined therapy for COVID-19 in immunocompromised patients: a promising approach against viral persistence and drug resistance. BMC Infect Dis 2025; 25:616. [PMID: 40295963 PMCID: PMC12036177 DOI: 10.1186/s12879-025-11012-3] [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: 11/07/2024] [Accepted: 04/18/2025] [Indexed: 04/30/2025] Open
Abstract
Immunocompromised (IC) patients face significant challenges in managing COVID-19 due to their heightened susceptibility to severe illness, persistent infections, and the potential development of drug resistance. Studies indicate that IC patients, particularly those with hematologic malignancies (HM), hematopoietic stem cell transplants (HSCTR), or solid organ transplants (SOTR), experience higher mortality rates and worse outcomes compared to the general population, even post-vaccination. The persistence of the virus in these patients, combined with its rapid mutation, further complicates treatment. Recent evidence supports the use of combined neutralizing monoclonal antibodies (mAbs) and direct-acting antivirals (DAAs) as a more effective approach to viral clearance, reducing mortality, and preventing relapses. However, the rise of resistant variants, especially to mAbs, and concerns about the safety of prolonged or intensive therapies pose ongoing challenges. Monotherapies often fail short to address these issues, highlighting the need for early combined therapy (ECT) with mAbs and DAAs. ECT has shown promise in managing COVID-19 in IC individuals by targeting multiple stages of the viral lifecycle, reducing viral load, and clearing infections at earlier stages, which helps mitigate the risks of severe disease and drug resistance. Continued research is essential to refine these treatment protocols, especially as the virus evolves. Although further studies are needed, current findings suggest that ECT may become the standard of care for managing COVID-19 in severely IC patients, offering better clinical outcomes and hindering viral persistence.
Collapse
Affiliation(s)
- Salvatore Rotundo
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.
- Infectious Diseases Unit, "San Giovanni di Dio" Hospital, Crotone, Italy.
| | - Francesca Serapide
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
- Infectious and Tropical Disease Unit, "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| | - Lavinia Berardelli
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
- Infectious and Tropical Diseases Unit, "Annunziata" Hospital, Cosenza, Italy
| | - Sara Palma Gullì
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Simona Mongiardi
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Maria Teresa Tassone
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Enrico Maria Trecarichi
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
- Department of Life Sciences, Health and Healthcare Professions, Link Campus University, Rome, Italy
| | - Alessandro Russo
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
- Infectious and Tropical Disease Unit, "Renato Dulbecco" Teaching Hospital, Catanzaro, Italy
| |
Collapse
|
2
|
Galota F, Marcheselli S, De Biasi S, Gibellini L, Vitetta F, Fiore A, Smolik K, De Napoli G, Cardi M, Cossarizza A, Ferraro D. Impact of High-Efficacy Therapies for Multiple Sclerosis on B Cells. Cells 2025; 14:606. [PMID: 40277931 PMCID: PMC12025603 DOI: 10.3390/cells14080606] [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: 02/27/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative autoimmune disorder of the central nervous system characterized by demyelination and neurodegeneration. Traditionally considered a T-cell-mediated disease, the crucial role of B lymphocytes in its pathogenesis, through different mechanisms contributing to inflammation and autoreactivity, is increasingly recognized. The risk of long-term disability in MS patients can be reduced by an early treatment initiation, in particular with high-efficacy therapies. The aim of this review is to provide an overview of the mechanisms of action of high-efficacy therapies for MS, with a focus on their impact on B cells and how this contributes to the drugs' efficacy and safety profiles. Anti-CD20 monoclonal antibodies, Alemtuzumab, Cladribine and sequestering therapies encompassing Natalizumab and S1P receptors modulators will be discussed and emerging therapies, including Bruton's Tyrosine Kinase inhibitors, will be presented.
Collapse
Affiliation(s)
- Federica Galota
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (F.G.); (S.M.); (K.S.); (M.C.)
| | - Simone Marcheselli
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (F.G.); (S.M.); (K.S.); (M.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, 41125 Modena, Italy; (S.D.B.); (L.G.); (A.C.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, 41125 Modena, Italy; (S.D.B.); (L.G.); (A.C.)
| | - Francesca Vitetta
- Neurology Unit, Department of Neurosciences, Ospedale Civile di Baggiovara, Azienda Ospedaliero-Universitaria di Modena, 41126 Modena, Italy; (F.V.); (A.F.)
| | - Alessia Fiore
- Neurology Unit, Department of Neurosciences, Ospedale Civile di Baggiovara, Azienda Ospedaliero-Universitaria di Modena, 41126 Modena, Italy; (F.V.); (A.F.)
| | - Krzysztof Smolik
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (F.G.); (S.M.); (K.S.); (M.C.)
| | - Giulia De Napoli
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (F.G.); (S.M.); (K.S.); (M.C.)
| | - Martina Cardi
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (F.G.); (S.M.); (K.S.); (M.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, 41125 Modena, Italy; (S.D.B.); (L.G.); (A.C.)
- National Institute for Cardiovascular Research, 40126 Bologna, Italy
| | - Diana Ferraro
- Department of Biomedical, Metabolic and Neurosciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (F.G.); (S.M.); (K.S.); (M.C.)
- Neurology Unit, Department of Neurosciences, Ospedale Civile di Baggiovara, Azienda Ospedaliero-Universitaria di Modena, 41126 Modena, Italy; (F.V.); (A.F.)
| |
Collapse
|
3
|
Hoechstetter MA, Hollwich E, Illner D, Pham T, von Bergwelt‐Baildon M, Dreyling M, Wendtner C. SARS-COV-2 Pre-Exposure Prophylaxis With Tixagevimab-Cilgavimab in Haematological, Immunocompromised Patients in the Omicron Era. Eur J Haematol 2025; 114:690-699. [PMID: 39757858 PMCID: PMC11880995 DOI: 10.1111/ejh.14377] [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: 09/23/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Pre-exposure prophylaxis with tixagevimab-cilgavimab significantly reduced severe COVID-19 outcomes in high-risk individuals during the pre-Omicron era (PROVENT trial). However, efficacy in patients with haematological malignancies (HM) was underreported. The rapid emergence of Omicron strains in 2021 showed reduced neutralizing activity in preclinical data, but real-world data remains limited due to short follow-up. PATIENTS AND METHODS We aimed to evaluate the effectiveness and safety of tixagevimab-cilgavimab in 86 HM patients during the early Omicron wave, including the BA.2, BA.5, and XBB.1 sublineages. These patients received PrEP between February and August 2022 due to impaired vaccine response (72%) and B-cell depletion (46.5%). They were followed prospectively until April 2023, with a median follow-up of 297 days. RESULTS Breakthrough SARS-CoV-2 infections occurred in 32.6% of patients, with 22.1% within six months. Infections within six months were milder and shorter. B-cell depletion within six (p = 0.035) and twelve months (p = 0.016) was identified as risk factor for breakthrough infections. No new safety events were reported. CONCLUSION Our data showed that tixagevimab-cilgavimab prophylaxis effectively reduces severe COVID-19 outcomes in patients with HM, particularly within the first six months, even during the Omicron era. However, those with recent B-cell depletion (within 12 months) remained at high risk.
Collapse
Affiliation(s)
- Manuela A. Hoechstetter
- Department of Haematology, Oncology, Immunology, Palliative Medicine, Infectious Diseases and Tropical MedicineMuenchen Klinik SchwabingMunichGermany
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University HospitalLudwig‐Maximilians UniversityMunichGermany
| | - Eva‐Maria Hollwich
- Department of Haematology, Oncology, Immunology, Palliative Medicine, Infectious Diseases and Tropical MedicineMuenchen Klinik SchwabingMunichGermany
| | - Doris Illner
- Department of Haematology, Oncology, Immunology, Palliative Medicine, Infectious Diseases and Tropical MedicineMuenchen Klinik SchwabingMunichGermany
| | - Thu‐Trang Pham
- Department of Haematology, Oncology, Immunology, Palliative Medicine, Infectious Diseases and Tropical MedicineMuenchen Klinik SchwabingMunichGermany
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University HospitalLudwig‐Maximilians UniversityMunichGermany
| | - Michael von Bergwelt‐Baildon
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University HospitalLudwig‐Maximilians UniversityMunichGermany
- German Cancer Consortium (DKTK), partner Site MunichA Partnership Between the DKFZ Heidelberg and the University Hospital of the LMUMunichGermany
| | - Martin Dreyling
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University HospitalLudwig‐Maximilians UniversityMunichGermany
| | - Clemens‐Martin Wendtner
- Department of Haematology, Oncology, Immunology, Palliative Medicine, Infectious Diseases and Tropical MedicineMuenchen Klinik SchwabingMunichGermany
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University HospitalLudwig‐Maximilians UniversityMunichGermany
| |
Collapse
|
4
|
Amdisen L, Pedersen L, Abildgaard N, Benn CS, Cronin‐Fenton D, Sørup S. Influenza vaccine effectiveness in immunocompromised patients with cancer: A Danish nationwide register-based cohort study. Cancer 2025; 131:e35574. [PMID: 39306693 PMCID: PMC11694166 DOI: 10.1002/cncr.35574] [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: 01/08/2024] [Revised: 07/02/2024] [Accepted: 07/16/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND Influenza vaccination is free of charge for Danish citizens with acquired immunodeficiency but recommendations do not specifically target patients with cancer. This study investigated whether influenza vaccination reduces the main outcome of overall mortality and the secondary outcomes of influenza requiring treatment, pneumonia, myocardial infarction, stroke, heart failure, and venous thromboembolism in patients with cancer. METHODS This was a register-based nationwide cohort study. Adjusted hazard ratios (aHRs) and 95% confidence intervals (95% CIs) for overall mortality and secondary outcomes were estimated using Cox proportional hazards models. Analyses were conducted separately for four subgroups: patients aged <65 years with solid tumors, patients aged ≥65 years with solid tumors, patients aged <65 years with hematological cancer, and patients aged ≥65 years with hematological cancer. RESULTS A total of 53,249 adult patients with solid tumors who received chemotherapy and 22,182 adult patients with hematological cancer were followed for up to five influenza seasons in the study period of 2007-2018. In the main analysis covering December-March, influenza vaccination was associated with reduced overall mortality in all four subgroups. The reduction was most pronounced in patients with hematological cancer aged <65 years (aHR, 0.66; 95% CI, 0.51-0.87) and smallest in patients with solid tumors aged <65 years (aHR, 0.91; 95% CI, 0.84-0.99). In sensitivity analyses covering January-March, the aHR was 0.87 (95% CI, 0.65-1.16) in patients with hematological cancer aged <65 years and 1.01 (95% CI, 0.92-1.10) in patients with solid tumors aged <65 years. Results for the secondary outcomes were inconclusive. CONCLUSIONS The results of this study cannot reject that influenza vaccination reduces overall mortality in immunocompromised patients with cancer. The results must be interpreted with caution because of potential unmeasured confounding, which can result in the overestimation of influenza vaccine effectiveness.
Collapse
Affiliation(s)
- Lau Amdisen
- Department of Clinical EpidemiologyDepartment of Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | - Lars Pedersen
- Department of Clinical EpidemiologyDepartment of Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | - Niels Abildgaard
- Hematology Research UnitDepartment of HematologyOdense University Hospital and Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
| | - Christine Stabell Benn
- Bandim Health ProjectOPENDepartment of Clinical ResearchUniversity of Southern Denmark and Odense University HospitalOdenseDenmark
- Danish Institute of Advanced ScienceUniversity of Southern DenmarkOdenseDenmark
| | - Deirdre Cronin‐Fenton
- Department of Clinical EpidemiologyDepartment of Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| | - Signe Sørup
- Department of Clinical EpidemiologyDepartment of Clinical MedicineAarhus University and Aarhus University HospitalAarhusDenmark
| |
Collapse
|
5
|
Silva-Pinto A, Abreu I, Martins A, Bastos J, Araújo J, Pinto R. Vaccination After Haematopoietic Stem Cell Transplant: A Review of the Literature and Proposed Vaccination Protocol. Vaccines (Basel) 2024; 12:1449. [PMID: 39772108 PMCID: PMC11680230 DOI: 10.3390/vaccines12121449] [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: 11/19/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Haematopoietic stem cell transplantation (HCT) induces profound immunosuppression, significantly increasing susceptibility to severe infections. This review examines vaccinations' necessity, timing, and efficacy post-HCT to reduce infection-related morbidity and mortality. It aims to provide a structured protocol aligned with international and national recommendations. Methods: A systematic review of current guidelines and studies was conducted to assess vaccination strategies in HCT recipients. The analysis included the timing of vaccine administration, factors influencing efficacy, and contraindications. Recommendations for pre- and post-transplant vaccination schedules were synthesised, specifically for graft-versus-host disease (GVHD), immunosuppressive therapy, and hypogammaglobulinemia. Results: Vaccination is essential as specific immunity is often lost after HCT. Inactivated vaccines are recommended to commence three months post-transplant, including influenza, COVID-19, and pneumococcal vaccines. Live attenuated vaccines remain contraindicated for at least two years post-transplant and in patients with ongoing GVHD or immunosuppressive therapy. Factors such as GVHD and immunosuppressive treatments significantly impact vaccine timing and efficacy. The review also underscores the importance of pre-transplant vaccinations and ensuring that patients' close contacts are adequately immunised to reduce transmission risks. Conclusions: Implementing a structured vaccination protocol post-HCT is critical to improving patient outcomes. Timely and effective vaccination strategies can mitigate infection risks while addressing individual patient factors such as GVHD and immunosuppression. This review highlights the need for tailored vaccination approaches to optimize immune reconstitution in HCT recipients.
Collapse
Affiliation(s)
- André Silva-Pinto
- Infectious Diseases Department, São João Hospital, 4200-319 Porto, Portugal; (I.A.); (A.M.)
- Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal; (J.B.); (J.A.); (R.P.)
| | - Isabel Abreu
- Infectious Diseases Department, São João Hospital, 4200-319 Porto, Portugal; (I.A.); (A.M.)
- Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal; (J.B.); (J.A.); (R.P.)
| | - António Martins
- Infectious Diseases Department, São João Hospital, 4200-319 Porto, Portugal; (I.A.); (A.M.)
- Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal; (J.B.); (J.A.); (R.P.)
| | - Juliana Bastos
- Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal; (J.B.); (J.A.); (R.P.)
- Haematology Department, São João Hospital, 4200-319 Porto, Portugal
| | - Joana Araújo
- Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal; (J.B.); (J.A.); (R.P.)
- Haematology Department, São João Hospital, 4200-319 Porto, Portugal
| | - Ricardo Pinto
- Faculty of Medicine, University of Porto, 4099-002 Porto, Portugal; (J.B.); (J.A.); (R.P.)
- Haematology Department, São João Hospital, 4200-319 Porto, Portugal
| |
Collapse
|
6
|
Choy J, Lewis KL. In the SARS-CoV-2 era, do we need to be afraid of obinutuzumab? Br J Haematol 2024; 205:2113-2115. [PMID: 39331693 PMCID: PMC11637727 DOI: 10.1111/bjh.19786] [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: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024]
Abstract
In their paper, Pinto et al. report findings from the Italian URBAN study; an ambispective, observational, multicentre study evaluating the safety and efficacy of obinutuzumab-based therapy for advanced stage follicular lymphoma (FL) in routine practice. The URBAN substudy reported here examined severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outcomes in 299 patients with treatment-naïve advanced stage FL, treated with obinutuzumab-based chemoimmunotherapy and maintenance. The study began enrolling in September 2019, continuing enrolment throughout the pandemic, with cut-off for the current analysis of 31 January 2022; thus, it provides unique insights into various pandemic phases, including the impact of administration of SARS-CoV-2 vaccination. Commentary on: Pinto et al. Exposure to obinutuzumab does not affect outcomes of SARS-CoV-2 infection in vaccinated patients with newly diagnosed advanced-stage follicular lymphoma. Br J Haematol 2024; 205:2219-2227.
Collapse
Affiliation(s)
- Joleen Choy
- Sir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
| | - Katharine L. Lewis
- Sir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
- University of Western AustraliaNedlandsWestern AustraliaAustralia
| |
Collapse
|
7
|
Kontandreopoulou CN, Solomou EE, Kolorizos E, Diamantopoulos PT. Vaccine challenges in CLL: a comprehensive exploration of efficacy of SARS-CoV-2 immunization for patients with chronic lymphocytic leukemia. Ann Hematol 2024; 103:4971-4980. [PMID: 39008060 DOI: 10.1007/s00277-024-05869-8] [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: 06/04/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by disease- and treatment-related immunosuppression. Patients with CLL comprise a vulnerable population to coronavirus disease 2019 (COVID-19), while the protective effect of COVID-19 vaccination remains uncertain.We conducted a systematic review to evaluate published data reporting response to COVID-19 vaccination in patients with CLL. The primary outcome was the rate of seropositivity after full primary vaccination, while secondary outcomes were rates of positive neutralizing antibodies, cellular responses, and adverse events. Response after booster doses of vaccination was also evaluated.Twenty-three studies of full primary vaccination (12 CLL-specific with 1747 patients, 11 with mixed hematologic diseases including 1044 patients with CLL) with a total of 2791 patients, and eight studies on booster doses with 389 patients were included in the analysis. The serologic response varied between studies with a median of 55%. Where reported, the median neutralizing antibody response rate was 61.2% and the cellular response rate was 44.2%. Poor serologic response was noted in patients under active treatment with anti-CD20 monoclonal antibodies, BCL2, and BTK inhibitors.The present review highlights the substantially impaired humoral and cellular response to COVID-19 vaccination in patients with CLL with patients under active treatment being the most vulnerable.
Collapse
Affiliation(s)
- Christina-Nefeli Kontandreopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elena E Solomou
- Department of Internal Medicine, University of Patras Medical School, Rion, Greece.
| | - Epaminondas Kolorizos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis T Diamantopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
8
|
Pinto A, Caltagirone M, Battista M, Gazzoli GC, Patti C, Pennese E, De Lorenzo S, Pavone V, Merli M, Chiarenza A, Gorgone AG, Piazza F, Puccini B, Noto A, Arcaini L, De Filippi R, Zinzani PL, Ferreri AJM, Ladetto M, Ferrari S, Gritti G. Exposure to obinutuzumab does not affect outcomes of SARS-CoV-2 infection in vaccinated patients with newly diagnosed advanced-stage follicular lymphoma. Br J Haematol 2024; 205:2219-2227. [PMID: 39039666 PMCID: PMC11637738 DOI: 10.1111/bjh.19661] [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: 04/11/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
URBAN is a multicentric, ambispective study evaluating the effectiveness and safety of obinutuzumab-based immuno-chemotherapy and maintenance in patients with untreated advanced follicular lymphoma (FL). The study began before the COVID-19 emergency declaration in Italy. It is currently ongoing for follow-up, and the enrolment timeline encompassed different stages of the pandemic, various vaccination roll-out phases and prevalence of SARS-CoV-2 variants. Outcomes of interest of the present sub-analysis included SARS-CoV-2 infection rates and COVID-19-related hospitalizations/deaths. At data cut-off, 86 (28.8%) and 213 patients (71.2%) were treated before and during/after the COVID-19 outbreak respectively; 294 (98.3%) completed the induction, 31 (10.4%) completed maintenance and 170 (56.9%) were still on maintenance. Overall, 245 patients (81.9%) received at least one SARS-CoV-2 vaccine dose: 13.5%, 31.4% and 55.1% received one, two and three doses respectively. We observed a substantial decrease in COVID-19-related mortality rates in pre- versus post-vaccination phases, along with a reduction in COVID-19-related outcomes due to the shift from alpha/delta to omicron variant predominance. No differences emerged between patients given maintenance or not, although the schedule was modified in 65% of cases. To our knowledge, URBAN represents the largest dataset of COVID-19-related outcomes in FL patients extensively exposed to obinutuzumab. ClinicalTrials.gov identifier: NCT04034056.
Collapse
MESH Headings
- Humans
- COVID-19/prevention & control
- COVID-19/mortality
- Lymphoma, Follicular/drug therapy
- Lymphoma, Follicular/mortality
- Male
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Female
- Middle Aged
- Aged
- SARS-CoV-2
- COVID-19 Vaccines/adverse effects
- COVID-19 Vaccines/administration & dosage
- Italy/epidemiology
- Adult
- Vaccination
- Aged, 80 and over
- Hospitalization/statistics & numerical data
- Neoplasm Staging
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Treatment Outcome
Collapse
Affiliation(s)
- A. Pinto
- Ematologia OncologicaIstituto Nazionale Tumori, Fondazione ‘G. Pascale’, IRCCSNaplesItaly
| | | | | | - G. C. Gazzoli
- International Patient Safety, Roche S.p.A.MonzaItaly
| | - C. Patti
- Ospedali Riuniti Villa Sofia – CervelloPalermoItaly
| | - E. Pennese
- Unità Operativa Complessa Ematologia Presidio Ospedaliero PescaraPescaraItaly
| | - S. De Lorenzo
- U.O.C. “Ematologia e TMO” AORN “S. G. Moscati”AvellinoItaly
| | - V. Pavone
- Unità Operativa Complessa di Ematologia e Trapianto di Cellule Staminali Emopoietiche, Azienda Ospedaliera Cardinale PanicoTricaseItaly
| | - M. Merli
- HematologyFondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - A. Chiarenza
- U.O.C. Ematologia Con TMO, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico – San Marco”CataniaItaly
| | | | - F. Piazza
- UOC Ematologia, Azienda Ospedaliero‐Universitaria di Padova, University of PadovaPadovaItaly
| | - B. Puccini
- EmatologiaAzienda Ospedaliero‐Universitaria CareggiFlorenceItaly
| | - A. Noto
- HematologyFondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - L. Arcaini
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo di PaviaUniversity of PaviaPaviaItaly
| | - R. De Filippi
- Dipartimento di Medicina Clinica e ChirurgiaScuola di Medicina, Università Federico IINaplesItaly
| | - P. L. Zinzani
- IRCCS Azienda Ospedaliero‐Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”BolognaItaly
- Dipartimento di Scienze Mediche e ChirurgicheUniversità di BolognaBolognaItaly
| | - A. J. M. Ferreri
- Lymphoma UnitIRCCS San Raffaele Scientific Institute and University Vita‐Salute San RaffaeleMilanItaly
| | - M. Ladetto
- Dipartimento di Medicina TraslazionaleUniversità del Piemonte Orientale SCDU Ematologia, AOU SS Antonio e Biagio e Cesare ArrigoAlessandriaItaly
| | - S. Ferrari
- UOC Ematologia Ospedale Papa Giovanni XXIIIBergamoItaly
| | - G. Gritti
- UOC Ematologia Ospedale Papa Giovanni XXIIIBergamoItaly
| |
Collapse
|
9
|
Tsoutsoukis M, Anthias C, Easdale S, Nicholson E. Re-vaccination against SARS-CoV-2 in allogeneic HSCT patients: Repeated primary vaccine doses increase seroconversion rates. Br J Haematol 2024; 205:1720-1726. [PMID: 39313909 DOI: 10.1111/bjh.19780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
Patients with haematological malignancies often exhibit reduced antibody responses to severe acute respiratory syndrome-related coronavirus vaccines, especially those who have undergone allogeneic haematopoietic stem cell transplantation (HSCT). Limited data exist on vaccine efficacy in this group. In a retrospective analysis of 75 post-HSCT patients, we assessed serologic responses to one to four doses of Pfizer-BioNTech (PB), AstraZeneca (AZ) or Moderna (MU) vaccines within 2 years post-transplant. Seroconversion rates were 50.7%, 78%, 79% and 83% after the first to fourth doses respectively. The median time from allograft to first re-vaccination was 145 days (range 79-700). Failure to respond to the first vaccine dose was linked to acute GVHD (p = 0.042) and rituximab treatment within 12 months (p = 0.019). A trend was observed with chronic GVHD and seroconversion failure after the second (p = 0.07) and third (p = 0.09) doses. Patients vaccinated before HSCT showed better antibody responses post-transplant (p = 0.019). Coronavirus disease 2019 incidence was 16%, with 17% hospitalized and one death (8%). Despite low initial seroconversion rates post-HSCT, antibody responses improved after the second dose. Early full re-vaccination and boosters post-HSCT are recommended to reduce mortality. Rituximab use and active GVHD were identified as risk factors, warranting further investigation.
Collapse
Affiliation(s)
- Marios Tsoutsoukis
- The Royal Marsden NHS Hospital, Sutton, UK
- United Lincolnshire Hospitals NHS Trust, Lincoln, UK
| | | | | | | |
Collapse
|
10
|
Gallais Sérézal I, Spehner L, Kroemer M, Bourezane I, Meaux-Ruault N, Prati C, Pastissier A, Lodovichetti J, Tiberghien P, Aubin F. Timing is essential: Humoral and cellular responses to SARS-CoV-2 vaccination in a cohort of patients with auto-immune diseases treated with rituximab. Heliyon 2024; 10:e38043. [PMID: 39328554 PMCID: PMC11425176 DOI: 10.1016/j.heliyon.2024.e38043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Rituximab (RTX), an anti CD20 monoclonal antibody, is now a gold standard treatment for several auto-immune and chronic inflammatory diseases. Receiving RTX exposes patients to more severe infections as vaccinations become virtually inefficient in terms of B cell responses. During the COVID-19 crisis, RTX-exposed patients exhibited more severe forms of the disease, and in some cases, the introduction of RTX was delayed or avoided to protect patients as much as possible against SARS-CoV-2 infections. We retrospectively collected cellular and humoral responses from thirteen patients with dermatological and rheumatological autoimmune diseases who had been vaccinated after receiving RTX. Memory T cells subsets from patients that exposed to RTX showed few differences when compared to a cohort of healthy donors. The IFNᵧ ELISpot assay using SARS-CoV-Prot_S1 showed that eight patients exhibited a positive response that was neither correlated to the time between RTX infusion and the sampling nor to the time between RTX and the vaccination. Conversely, analysis of the SARS-CoV-2 serology showed a clearly lower binding antibody units per mL in case of recent RTX infusion. The safe threshold forconsistently positive serology was to vaccinate at least 300 days after RTX infusion (p = 0.02). Our data illustrate the difficulty in obtaining a satisfactory response to vaccination after RTX treatment within almost a year after the latest infusion, and emphasize the need to better evaluate the risk of relapses in auto-immune diseases before administering RTX in order to maintain RTX only in patients whose medical situation requires it.
Collapse
Affiliation(s)
- Irène Gallais Sérézal
- Department of Dermatology, Besançon University Hospital, Besançon, France
- Université de Franche-Comté, EFS, INSERM UMR RIGHT, Besançon, France
| | - Laurie Spehner
- Université de Franche-Comté, EFS, INSERM UMR RIGHT, Besançon, France
- Inserm CIC-1431, CHU Besançon, Besançon, France
| | - Marie Kroemer
- Université de Franche-Comté, EFS, INSERM UMR RIGHT, Besançon, France
- Department of Pharmacy, Besançon University Hospital, Besançon, France
| | - Inès Bourezane
- Université de Franche-Comté, EFS, INSERM UMR RIGHT, Besançon, France
- Department of Pharmacy, Besançon University Hospital, Besançon, France
| | - Nadine Meaux-Ruault
- Department of Internal Medicine, Besançon University Hospital, Besançon, France
| | - Clément Prati
- Department of Rheumatology, Besançon University Hospital, Besançon, France
| | - Andréa Pastissier
- Department of Internal Medicine, Besançon University Hospital, Besançon, France
| | | | - Pierre Tiberghien
- Université de Franche-Comté, EFS, INSERM UMR RIGHT, Besançon, France
- Etablissement Francais Du Sang, La Plaine-Saint Denis, Saint-Denis, France
| | - François Aubin
- Department of Dermatology, Besançon University Hospital, Besançon, France
- Université de Franche-Comté, EFS, INSERM UMR RIGHT, Besançon, France
| |
Collapse
|
11
|
Sherman AC, van Haren SD, Borberg E, Swank Z, Aleissa M, Tong A, Rooks R, Kanwal U, Levine H, Yates B, Izaguirre N, Ryff K, Thomas S, Parisi L, Li X, Walt DR, Levy O, Walsh SR, Issa NC, Baden LR. Heterologous SARS-CoV-2 booster vaccine for individuals with hematological malignancies after a primary SARS-CoV-2 mRNA vaccine series. Vaccine 2024; 42:126054. [PMID: 38862310 DOI: 10.1016/j.vaccine.2024.05.081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/13/2024]
Abstract
Heterologous COVID-19 vaccine boosters have not been evaluated for patients with hematological malignancies. A Novavax booster was administered for 56 individuals with hematological malignancies who had received a primary COVID-19 series and prior boosters with mRNA vaccines only. Blood specimens were obtained at baseline (pre-vaccine), 28 days, and 168 days after vaccination with the Novavax booster. The median fold change of anti-Spike IgG was 1.02 (IQR 0.79, 1.3) between baseline and Day 28. Circulating Spike protein-specific B cells increased 1.4-fold at Day 28 (p < 0.05). Increases in antibody and T cell responses were modest without significance, with a waning of humoral and cellular responses at 168 days after vaccination.
Collapse
Affiliation(s)
- Amy C Sherman
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA; Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Ella Borberg
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Zoe Swank
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Muneerah Aleissa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA; Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Alexandra Tong
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Rebecca Rooks
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA; Emory School of Medicine, Atlanta, GA, USA
| | - Urwah Kanwal
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Hannah Levine
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Bridget Yates
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Natalie Izaguirre
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Kevin Ryff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Sanya Thomas
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Lindsey Parisi
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Xiaofang Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - David R Walt
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Stephen R Walsh
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Nicolas C Issa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Kavikondala S, Haeussler K, Wang X, Spellman A, Bausch-Jurken MT, Sharma P, Amiri M, Krivelyova A, Vats S, Nassim M, Kumar N, Van de Velde N. Immunogenicity of mRNA-1273 and BNT162b2 in Immunocompromised Patients: Systematic Review and Meta-analysis Using GRADE. Infect Dis Ther 2024; 13:1419-1438. [PMID: 38802704 PMCID: PMC11219657 DOI: 10.1007/s40121-024-00987-2] [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: 03/14/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Abstract
INTRODUCTION Immunocompromised (IC) patients mount poor immune responses to vaccination. Higher-dose coronavirus disease 2019 (COVID-19) vaccines may offer increased immunogenicity. METHODS A pairwise meta-analysis of 98 studies reporting comparisons of mRNA-1273 (50 or 100 mcg/dose) and BNT162b2 (30 mcg/dose) in IC adults was performed. Outcomes were seroconversion, total and neutralizing antibody titers, and cellular immune responses. RESULTS mRNA-1273 was associated with a significantly higher seroconversion likelihood [relative risk, 1.11 (95% CI, 1.08, 1.14); P < 0.0001; I2 = 66.8%] and higher total antibody titers [relative increase, 50.45% (95% CI, 34.63%, 66.28%); P < 0.0001; I2 = 89.5%] versus BNT162b2. mRNA-1273 elicited higher but statistically nonsignificant relative increases in neutralizing antibody titers and cellular immune responses versus BNT162b2. CONCLUSION Higher-dose mRNA-1273 had increased immunogenicity versus BNT162b2 in IC patients.
Collapse
|
13
|
Merli M, Costantini A, Tafuri S, Bavaro DF, Minoia C, Meli E, Luminari S, Gini G. Management of vaccinations in patients with non-Hodgkin lymphoma. Br J Haematol 2024; 204:1617-1634. [PMID: 38532527 DOI: 10.1111/bjh.19422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/07/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Vaccinations are fundamental tools in preventing infectious diseases, especially in immunocompromised patients like those affected by non-Hodgkin lymphomas (NHLs). The COVID-19 pandemic made clinicians increasingly aware of the importance of vaccinations in preventing potential life-threatening SARS-CoV-2-related complications in NHL patients. However, several studies have confirmed a significant reduction in vaccine-induced immune responses after anti-CD20 monoclonal antibody treatment, thus underscoring the need for refined immunization strategies in NHL patients. In this review, we summarize the existing data about COVID-19 and other vaccine's efficacy in patients with NHL and propose multidisciplinary team-based recommendations for the management of vaccines in this specific group of patients.
Collapse
Affiliation(s)
- Michele Merli
- Division of Hematology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Costantini
- Clinical Immunology Unit, Azienda Ospedaliero Universitaria delle Marche - Università Politecnica delle Marche, Ancona, Italy
| | - Silvio Tafuri
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Davide Fiore Bavaro
- Department of Biomedical Sciences and Human Oncology, Clinic of Infectious Diseases, Aldo Moro University of Bari, Bari, Italy
| | - Carla Minoia
- Hematology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Erika Meli
- Division of Hematology, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | - Stefano Luminari
- Hematology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Surgical Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Guido Gini
- Clinic of Hematology, Azienda Ospedaliero Universitaria Delle Marche - Università Politecnica Delle Marche, Ancona, Italy
| |
Collapse
|
14
|
New J, Cham J, Smith L, Puglisi L, Huynh T, Kurian S, Bagsic S, Fielding R, Hong L, Reddy P, Eum KS, Martin A, Barrick B, Marsh C, Quigley M, Nicholson LJ, Pandey AC. Effects of antineoplastic and immunomodulating agents on postvaccination SARS-CoV-2 breakthrough infections, antibody response, and serological cytokine profile. J Immunother Cancer 2024; 12:e008233. [PMID: 38296596 PMCID: PMC10831464 DOI: 10.1136/jitc-2023-008233] [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] [Accepted: 12/08/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Despite immunization, patients on antineoplastic and immunomodulating agents have a heightened risk of COVID-19 infection. However, accurately attributing this risk to specific medications remains challenging. METHODS An observational cohort study from December 11, 2020 to September 22, 2022, within a large healthcare system in San Diego, California, USA was designed to identify medications associated with greatest risk of postimmunization SARS-CoV-2 infection. Adults prescribed WHO Anatomical Therapeutic Chemical (ATC) classified antineoplastic and immunomodulating medications were matched (by age, sex, race, and number of immunizations) with control patients not prescribed these medications yielding a population of 26 724 patients for analysis. From this population, 218 blood samples were collected from an enrolled subset to assess serological response and cytokine profile in relation to immunization. RESULTS Prescription of WHO ATC classified antineoplastic and immunomodulatory agents was associated with elevated postimmunization SARS-CoV-2 infection risk (HR 1.50, 95% CI 1.38 to 1.63). While multiple immunization doses demonstrated a decreased association with postimmunization SARS-CoV-2 infection risk, antineoplastic and immunomodulatory treated patients with four doses remained at heightened risk (HR 1.23, 95% CI 1.06 to 1.43). Risk variation was identified among medication subclasses, with PD-1/PD-L1 inhibiting monoclonal antibodies, calcineurin inhibitors, and CD20 monoclonal antibody inhibitors identified to associate with increased risk of postimmunization SARS-CoV-2 infection. Antineoplastic and immunomodulatory treated patients also displayed a reduced IgG antibody response to SARS-CoV-2 epitopes alongside a unique serum cytokine profile. CONCLUSIONS Antineoplastic and immunomodulating medications associate with an elevated risk of postimmunization SARS-CoV-2 infection in a drug-specific manner. This comprehensive, unbiased analysis of all WHO ATC classified antineoplastic and immunomodulating medications identifies medications associated with greatest risk. These findings are crucial in guiding and refining vaccination strategies for patients prescribed these treatments, ensuring optimized protection for this susceptible population in future COVID-19 variant surges and potentially for other RNA immunization targets.
Collapse
Affiliation(s)
- Jacob New
- Medicine, Scripps Health, La Jolla, California, USA
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Jason Cham
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Lana Smith
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Leah Puglisi
- Medicine, Scripps Health, La Jolla, California, USA
| | - Tridu Huynh
- Scripps Research Translational Institute, La Jolla, California, USA
- Division of Hematology/Oncology, University of California, La Jolla, California, USA
| | - Sunil Kurian
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | | | - Russel Fielding
- Strategy & Planning, Scripps Health, La Jolla, California, USA
| | - Lee Hong
- Medicine, Scripps Health, La Jolla, California, USA
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Priya Reddy
- Medicine, Scripps Health, La Jolla, California, USA
| | - Ki Suk Eum
- Medicine, Scripps Health, La Jolla, California, USA
- Rheumatology, Veterans Administration Pacific Islands Healthcare System, Honolulu, Hawaii, USA
| | - Allison Martin
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | - Bethany Barrick
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | - Christopher Marsh
- Scripps Organ Transplantation Research & Biorepository, Scripps Health, La Jolla, California, USA
| | | | - Laura J Nicholson
- Medicine, Scripps Health, La Jolla, California, USA
- Scripps Research Translational Institute, La Jolla, California, USA
| | - Amitabh C Pandey
- Scripps Research Translational Institute, La Jolla, California, USA
- Medicine, Section of Cardiology, Tulane University, New Orleans, Louisiana, USA
- Medicine, Southeast Veterans Health Care System, New Orleans, Louisiana, USA
| |
Collapse
|
15
|
Hillyer A, Quint A, Ghassemian A, Joh-Carnella N, Knauer MJ, Dawd D, Lazo-Langner A, Mangel J, Lam S, Abdoh H, Xenocostas A, Deotare U, Saini L, Foster C, Louzada M, Ho J, Chin-Yee I, Phua CW. Serologic Response to Vaccine for COVID-19 in Patients with Hematologic Malignancy: A Prospective Cohort Study. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024:S2152-2650(24)00021-1. [PMID: 38336492 DOI: 10.1016/j.clml.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Patients with hematological cancers have increased COVID-19 morbidity and mortality, and these patients show attenuated vaccine responses. This study aimed to characterize the longitudinal humoral immune responses to COVID-19 vaccination in patients with hematological malignancies. PATIENTS AND METHODS We conducted a prospective cohort study, collecting samples from March 2021 to July 2022, from patients seen at a cancer treatment center in London, Ontario, Canada, who met the following eligibility criteria: age ≥18 years, diagnosed with a hematological malignancy, recipient of a COVID-19 vaccine during the study period, and able to provide informed consent. RESULTS Median anti-S titers (MST) were 0.0, 64.0, and 680.5 U/mL following first (V1), second (V2), and third (V3) vaccine doses, respectively. Patients with lymphoid malignancies' response to vaccination was attenuated compared to myeloid malignancy patients after V2 and V3 (P < .001, P < .01). Active treatment was associated with lower antibody titers (MST 10) compared to treatment 12-24 months (MST 465, P = .04367) and >24 months (MST 1660.5, P = .0025) prior to vaccination. V3 significantly increased antibody titers compared to V2 for patients less than 3 months from treatment. Increasing age was associated with smaller antibody response following V2 (P < .05), but not following V3. Patients receiving anti-CD20 therapy did not demonstrate increased antibody titer levels after V3 (V2 MST 0, V3 MST 0; P > .05). CONCLUSION We report an attenuated serologic response to COVID-19 vaccination in our study population of patients with hematological malignancy. The immune response to vaccination was affected by patient age, diagnosis, treatment, and timing of treatment exposure.
Collapse
Affiliation(s)
| | | | - Artin Ghassemian
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | | | - Michael J Knauer
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Danny Dawd
- Schulich School of Medicine and Dentistry, Western University, London ON
| | - Alejandro Lazo-Langner
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Joy Mangel
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Selay Lam
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Husam Abdoh
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Anargyros Xenocostas
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Uday Deotare
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Lalit Saini
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Cheryl Foster
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Martha Louzada
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Jenny Ho
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Ian Chin-Yee
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON
| | - Chai W Phua
- Division of Hematology and Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON.
| |
Collapse
|
16
|
Priddey A, Chen-Xu MXH, Cooper DJ, MacMillan S, Meisl G, Xu CK, Hosmillo M, Goodfellow IG, Kollyfas R, Doffinger R, Bradley JR, Mohorianu II, Jones R, Knowles TPJ, Smith R, Kosmoliaptsis V. Microfluidic antibody profiling after repeated SARS-CoV-2 vaccination links antibody affinity and concentration to impaired immunity and variant escape in patients on anti-CD20 therapy. Front Immunol 2024; 14:1296148. [PMID: 38259440 PMCID: PMC10800570 DOI: 10.3389/fimmu.2023.1296148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Background Patients with autoimmune/inflammatory conditions on anti-CD20 therapies, such as rituximab, have suboptimal humoral responses to vaccination and are vulnerable to poorer clinical outcomes following SARS-CoV-2 infection. We aimed to examine how the fundamental parameters of antibody responses, namely, affinity and concentration, shape the quality of humoral immunity after vaccination in these patients. Methods We performed in-depth antibody characterisation in sera collected 4 to 6 weeks after each of three vaccine doses to wild-type (WT) SARS-CoV-2 in rituximab-treated primary vasculitis patients (n = 14) using Luminex and pseudovirus neutralisation assays, whereas we used a novel microfluidic-based immunoassay to quantify polyclonal antibody affinity and concentration against both WT and Omicron (B.1.1.529) variants. We performed comparative antibody profiling at equivalent timepoints in healthy individuals after three antigenic exposures to WT SARS-CoV-2 (one infection and two vaccinations; n = 15) and in convalescent patients after WT SARS-CoV-2 infection (n = 30). Results Rituximab-treated patients had lower antibody levels and neutralisation titres against both WT and Omicron SARS-CoV-2 variants compared to healthy individuals. Neutralisation capacity was weaker against Omicron versus WT both in rituximab-treated patients and in healthy individuals. In the rituximab cohort, this was driven by lower antibody affinity against Omicron versus WT [median (range) KD: 21.6 (9.7-38.8) nM vs. 4.6 (2.3-44.8) nM, p = 0.0004]. By contrast, healthy individuals with hybrid immunity produced a broader antibody response, a subset of which recognised Omicron with higher affinity than antibodies in rituximab-treated patients [median (range) KD: 1.05 (0.45-1.84) nM vs. 20.25 (13.2-38.8) nM, p = 0.0002], underpinning the stronger serum neutralisation capacity against Omicron in the former group. Rituximab-treated patients had similar anti-WT antibody levels and neutralisation titres to unvaccinated convalescent individuals, despite two more exposures to SARS-CoV-2 antigen. Temporal profiling of the antibody response showed evidence of affinity maturation in healthy convalescent patients after a single SARS-CoV-2 infection, which was not observed in rituximab-treated patients, despite repeated vaccination. Discussion Our results enrich previous observations of impaired humoral immune responses to SARS-CoV-2 in rituximab-treated patients and highlight the significance of quantitative assessment of serum antibody affinity and concentration in monitoring anti-viral immunity, viral escape, and the evolution of the humoral response.
Collapse
Affiliation(s)
- Ashley Priddey
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Michael Xin Hua Chen-Xu
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel James Cooper
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Serena MacMillan
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Catherine K. Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Myra Hosmillo
- Department of Pathology, Division of Virology, University of Cambridge, Cambridge, United Kingdom
| | - Ian G. Goodfellow
- Department of Pathology, Division of Virology, University of Cambridge, Cambridge, United Kingdom
| | - Rafael Kollyfas
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - John R. Bradley
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Irina I. Mohorianu
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rachel Jones
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Tuomas P. J. Knowles
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - Rona Smith
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge and the NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| |
Collapse
|
17
|
Vanni A, Salvati L, Mazzoni A, Lamacchia G, Capone M, Francalanci S, Kiros ST, Cosmi L, Puccini B, Ciceri M, Sordi B, Rossolini GM, Annunziato F, Maggi L, Liotta F. Bendamustine impairs humoral but not cellular immunity to SARS-CoV-2 vaccination in rituximab-treated B-cell lymphoma-affected patients. Front Immunol 2023; 14:1322594. [PMID: 38106404 PMCID: PMC10722279 DOI: 10.3389/fimmu.2023.1322594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
Background Patients with B-cell lymphoma are a fragile category of subjects, particularly exposed to infections and characterized by an impaired vaccination response due to the disease itself and, even more, to the chemotherapy regimen. For this reason, extensive knowledge of the immune response status of these subjects is of fundamental importance to obtain possible indications for a tailored immunization strategy. Methods We enrolled two cohorts of patients with B-cell lymphoma under rituximab treatment or 3-24 months after treatment. In all patients, we evaluated both humoral and cellular immunological memory toward SARS-CoV-2, after standard vaccination and upon one booster dose. Results We observed no Spike-specific IgG production in patients (n = 25) under anti-CD20 treatment, whereas patients (n = 16) vaccinated after the completion of chemotherapy showed a higher humoral response. Evaluating SARS-CoV-2-specific T-cell response, we found that patients in both cohorts had developed robust cellular immunity after vaccination. Of the 21 patients (51%) that experienced a breakthrough SARS-CoV-2 infection, only six patients developed severe disease. Interestingly, these six patients had all been treated with rituximab plus bendamustine. Notably, we observed that Spike-specific IgG levels in patients treated with rituximab plus bendamustine were absent or lower compared with those in patients treated with rituximab plus other chemotherapy, whereas Spike-specific T-cell response was not different based on chemotherapy regiment. Discussion Our results show that, in patients with B-cell lymphoma under rituximab therapy, anti-SARS-CoV-2 mRNA vaccination induces a weak or absent humoral response but a consistent T-cell response. In addition, chemotherapy regimens with bendamustine further reduce patients' ability to mount a Spike-specific humoral response even after a long time period from chemotherapy discontinuation. These results provide evidence that different chemotherapeutics display different immunosuppressive properties that could be taken in to account in the choice of the right drug regimen for the right patient. Moreover, they question whether immunocompromised patients, particularly those treated with bendamustine, need interventions to improve vaccine-induced immune response.
Collapse
Affiliation(s)
- Anna Vanni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Giulia Lamacchia
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Manuela Capone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Stefania Francalanci
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Seble Tekle Kiros
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | | | - Manuel Ciceri
- Hematology Unit, Careggi University Hospital, Florence, Italy
| | - Benedetta Sordi
- Hematology Unit, Careggi University Hospital, Florence, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunology and Cell Therapy Unit, Careggi University Hospital, Florence, Italy
| |
Collapse
|
18
|
Gutierrez A, Alonso A, Garcia-Recio M, Perez S, Garcia-Maño L, Martinez-Serra J, Ros T, Garcia-Gasalla M, Ferrer J, Vögler O, Alemany R, Salar A, Sampol A, Bento L. Analysis of vaccine responses after anti-CD20 maintenance in B-cell lymphoma in the Balearic Islands. A single reference center experience. Front Immunol 2023; 14:1267485. [PMID: 38022668 PMCID: PMC10646481 DOI: 10.3389/fimmu.2023.1267485] [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: 07/26/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The use of maintenance approaches with anti-CD20 monoclonal antibodies has improved the outcomes of B-cell indolent lymphomas but may lead to significant peripheral B-cell depletion. This depletion can potentially hinder the serological response to neoantigens. Methods Our objective was to analyze the effect of anti-CD20 maintenance therapy in a reliable model of response to neoantigens: SARS-CoV-2 vaccine responses and the incidence/severity ofCOVID-19 in a reference hospital. Results In our series (n=118), the rate of vaccination failures was 31%. Through ROC curve analysis, we determined a cutoff for SARS-CoV-2 vaccine serologic response at 24 months from the last anti-CD20 dose. The risk of severe COVID-19 was notably higher within the first 24months following the last anti-CD20 dose (52%) compared to after this period (just 18%) (p=0.007). In our survival analysis, neither vaccine response nor hypogammaglobulinemia significantly affected OS. While COVID-19 led to a modest mortality rate of 2.5%, this figure was comparable to the OS reported in the general immunocompetent population. However, most patients with hypogammaglobulinemia received intravenous immunoglobulin therapy and all were vaccinated. In conclusion, anti-CD20 maintenance therapy impairs serological responses to SARS-CoV-2 vaccines. Discussion We report for the first time that patients during maintenance therapy and up to 24 months after the last anti-CD20 dose are at a higher risk of vaccine failure and more severe cases of COVID-19. Nevertheless, with close monitoring, intravenous immunoglobulin supplementation or proper vaccination, the impact on survival due to the lack of serological response in this high-risk population can be mitigated, allowing for the benefits of anti-CD20 maintenance therapy, even in the presence of hypogammaglobulinemia.
Collapse
Affiliation(s)
- Antonio Gutierrez
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Aser Alonso
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Marta Garcia-Recio
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Sandra Perez
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Lucia Garcia-Maño
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jordi Martinez-Serra
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Teresa Ros
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Mercedes Garcia-Gasalla
- Service of Internal Medicine and Infecious Diseases, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Joana Ferrer
- Service of Immunology, University Hospital Son Espases, Palma, Spain
| | - Oliver Vögler
- Group of Advanced Therapies and Biomarkers in Clinical Oncology, Health Research Institute of the Balearic Islands (IdISBa), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Group of Clinical and Translational Research, Department of Biology, University of the Balearic Islands, Palma, Spain
| | - Regina Alemany
- Group of Advanced Therapies and Biomarkers in Clinical Oncology, Health Research Institute of the Balearic Islands (IdISBa), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands, Palma, Spain
- Group of Clinical and Translational Research, Department of Biology, University of the Balearic Islands, Palma, Spain
| | - Antonio Salar
- Service of Hematology , Hospital Clinico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Antonia Sampol
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Leyre Bento
- Service of Hematology, University Hospital Son Espases/Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| |
Collapse
|
19
|
Petri M, Joyce D, Haag K, Fava A, Goldman DW, Zhong D, Xiao S, Milstone A, Magder LS. Effect of Systemic Lupus Erythematosus and Immunosuppressive Agents on COVID-19 Vaccination Antibody Response. Arthritis Care Res (Hoboken) 2023; 75:1878-1885. [PMID: 36714913 PMCID: PMC10387122 DOI: 10.1002/acr.25094] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 01/03/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
OBJECTIVE The risk of COVID-19 infection is increased in patients with systemic lupus erythematosus (SLE) versus those without SLE. Some immunosuppressive medications increase COVID-19 infection and decrease the efficacy of vaccination. Consensus documents have suggested management strategies for handling immunosuppressive medications to increase vaccine efficacy, but the benefit of such strategies has not been proven. The current study was undertaken to determine the effect of immunosuppressive drugs on vaccine response in SLE. METHODS We collected information on COVID-19 infection, vaccination history, and COVID-19 antibodies in the Hopkins Lupus Cohort. A cohort of health care workers was used for comparison. Outcome measures included SARS-CoV-2 antibody IgG levels after vaccination over time in both cohorts and effect of immunosuppressive medications on postvaccination IgG levels in SLE patients. RESULTS The analysis was based on 365 observations from 334 different patients in the SLE cohort, and 2,235 observations from 1,887 different health care workers. SLE patients taking immunosuppressive medications had lower vaccine IgG levels than SLE patients who were not; but both groups had lower levels than health care workers. Holding mycophenolate for 1 week after vaccination increased postvaccine IgG levels significantly without leading to clinical flares. In multiple variable models, mycophenolate mofetil, tacrolimus, and belimumab all significantly reduced antibody response to vaccination. CONCLUSION SLE patients, regardless of background immunosuppressive therapy, had lower vaccine IgG levels than health care workers. Mycophenolate, tacrolimus, and belimumab significantly reduced IgG response to vaccination. Holding mycophenolate for 1 week improved vaccine efficacy, providing clinical benefit on vaccine response without leading to clinical flares.
Collapse
Affiliation(s)
- Michelle Petri
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Rheumatology
| | - Daniel Joyce
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Rheumatology
| | - Kristin Haag
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Rheumatology
| | - Andrea Fava
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Rheumatology
| | - Daniel W. Goldman
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Rheumatology
| | - Diana Zhong
- Johns Hopkins University School of Medicine, Department of Medicine, Division of Infectious Diseases
| | - Shaoming Xiao
- Johns Hopkins University School of Medicine, Department of Pediatrics. Division of Infectious Diseases
| | - Aaron Milstone
- Johns Hopkins University School of Medicine, Department of Pediatrics. Division of Infectious Diseases
| | - Laurence S. Magder
- University of Baltimore School of Medicine, Department of Epidemiology and Biostatistics
| |
Collapse
|
20
|
Koehm M, Klippstein M, Dauth S, Hallmann K, Kohmer N, Burkhardt H, Ciesek S, Geisslinger G, Rabenau HF, Behrens F. Impact of different classes of immune-modulating treatments on B cell-related and T cell-related immune response before and after COVID-19 booster vaccination in patients with immune-mediated diseases and primary immunodeficiency: a cohort study. RMD Open 2023; 9:e003094. [PMID: 37652553 PMCID: PMC10476126 DOI: 10.1136/rmdopen-2023-003094] [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: 02/21/2023] [Accepted: 06/23/2023] [Indexed: 09/02/2023] Open
Abstract
OBJECTIVES To evaluate the potential of immunosuppressed patients to mount B-cell and T-cell responses to COVID-19 booster vaccination (third vaccination). METHODS Patients with primary immunodeficiency (PID), immune-mediated inflammatory diseases (IMIDs) on CD20-depleting treatment with rituximab (RTX), or IMIDs treated with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) or biological disease-modifying antirheumatic drug (bDMARDs) were included and assessed before (baseline visit (BL)) and 2, 4 and 8 weeks after COVID-19 booster vaccination. Serum B-cell responses were assessed by antibody levels against SARS-CoV-2 spike protein (anti-spike IgG antibody (S-AB)) and a surrogate virus neutralisation test (sVNT). T-cell responses were assessed by an interferon gamma release assay (IGRA). RESULTS Fifty patients with PID (n=6), treated with RTX therapy (n=13), or treated with csDMARDs/bDMARDs (n=31) were included. At BL, anti-S-AB titres in PID and csDMARD/bDMARD-treated patients were low (although significantly higher than RTX patients); measures of B-cell-mediated response increased significantly after booster vaccination. In the RTX cohort, low BL anti-S-AB and sVNT values did not improve after booster vaccination, but patients had significantly elevated IGRA responses post booster vaccination compared with the other groups. csDMARD/bDMARD-treated patients showed the highest BL values in all three assays with greater increases in all parameters after booster vaccination compared with patients with PID. CONCLUSION Patients with IMID on therapeutic B-cell depletion have low anti-S-AB and sVNT values before and after booster vaccination but show significantly higher levels of IGRA compared with other immunosuppressed patients, suggesting an underlying mechanism attempting to compensate compromised humoral immunity by upregulating T-cell responsiveness. PID appears to have a stronger impact on antiviral immune response than csDMARD/bDMARD treatment.
Collapse
Affiliation(s)
- Michaela Koehm
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Maximilian Klippstein
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Stephanie Dauth
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Konstantin Hallmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Niko Kohmer
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Harald Burkhardt
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Sandra Ciesek
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- Virology, German Centre for Infection Research, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Pharmazentrum Frankfurt/ZAFES, Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany
| | - Holger F Rabenau
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Frank Behrens
- Department of Rheumatology, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
| |
Collapse
|
21
|
Otero-Romero S, Lebrun-Frénay C, Reyes S, Amato MP, Campins M, Farez M, Filippi M, Hacohen Y, Hemmer B, Juuti R, Magyari M, Oreja-Guevara C, Siva A, Vukusic S, Tintoré M. ECTRIMS/EAN consensus on vaccination in people with multiple sclerosis: Improving immunization strategies in the era of highly active immunotherapeutic drugs. Mult Scler 2023; 29:904-925. [PMID: 37293841 PMCID: PMC10338708 DOI: 10.1177/13524585231168043] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/30/2023] [Accepted: 03/19/2023] [Indexed: 06/10/2023]
Abstract
BACKGROUND With the new highly active drugs available for people with multiple sclerosis (pwMS), vaccination becomes an essential part of the risk management strategy. OBJECTIVE To develop a European evidence-based consensus for the vaccination strategy of pwMS who are candidates for disease-modifying therapies (DMTs). METHODS This work was conducted by a multidisciplinary working group using formal consensus methodology. Clinical questions (defined as population, interventions, and outcomes) considered all authorized DMTs and vaccines. A systematic literature search was conducted and quality of evidence was defined according to the Oxford Centre for Evidence-Based Medicine Levels of Evidence. The recommendations were formulated based on the quality of evidence and the risk-benefit balance. RESULTS Seven questions, encompassing vaccine safety, vaccine effectiveness, global vaccination strategy and vaccination in sub-populations (pediatric, pregnant women, elderly and international travelers) were considered. A narrative description of the evidence considering published studies, guidelines, and position statements is presented. A total of 53 recommendations were agreed by the working group after three rounds of consensus. CONCLUSION This first European consensus on vaccination in pwMS proposes the best vaccination strategy according to current evidence and expert knowledge, with the goal of homogenizing the immunization practices in pwMS.
Collapse
Affiliation(s)
- Susana Otero-Romero
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Barcelona Hospital, Barcelona, Spain Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| | | | - Saúl Reyes
- Fundación Santa Fe de Bogotá, Bogotá, Colombia School of Medicine, Universidad de los Andes, Bogotá, Colombia Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Maria Pia Amato
- Department NEUROFARBA, University of Florence, Florence, Italy IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Magda Campins
- Department of Preventive Medicine and Epidemiology, Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| | - Mauricio Farez
- Centro para la Investigación de Enfermedades Neuroinmunológicas (CIEN), FLENI, Buenos Aires, Argentina
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy Neurology Unit, Neurorehabilitation Unit, and Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University, Milan, Italy
| | - Yael Hacohen
- Department of Paediatric Neurology, Great Ormond Street Hospital for Children, London, UK Department of Neuroinflammation, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Bernhard Hemmer
- Department of Neurology, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Rosa Juuti
- Multiple Sclerosis International Federation, London, UK
| | - Melinda Magyari
- Department of Neurology, Danish Multiple Sclerosis Center and the Danish Multiple Sclerosis Registry, Rigshospitalet and University of Copenhagen, Copenhagen, Denmark
| | - Celia Oreja-Guevara
- Department of Neurology, Hospital Clínico San Carlos, IdISSC, Departamento de Medicina, Universidad Complutense, Madrid, Spain
| | - Aksel Siva
- Department of Neurology, School of Medicine, Istanbul University Cerrahpasa, Cerrahpasa, Istanbul, Turkey
| | - Sandra Vukusic
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuro-inflammation, Centre de Référence des Maladies Inflammatoires Rares du Cerveau et de la Moelle, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon, France
- Centre des Neurosciences de Lyon, Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
| | - Mar Tintoré
- Multiple Sclerosis Centre of Catalonia (Cemcat), Vall d’Hebron Barcelona Hospital, Barcelona, Spain
| |
Collapse
|
22
|
Kokogho A, Crowell TA, Aleissa M, Lupan AM, Davey S, Park Chang JB, Baden LR, Walsh SR, Sherman AC. SARS-CoV-2 Vaccine-Induced Immune Responses Among Hematopoietic Stem Cell Transplant Recipients. Open Forum Infect Dis 2023; 10:ofad349. [PMID: 37520415 PMCID: PMC10372870 DOI: 10.1093/ofid/ofad349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/06/2023] [Indexed: 08/01/2023] Open
Abstract
Background Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination reduces the risk and severity of coronavirus disease 2019 (COVID-19), several variables may impact the humoral response among patients undergoing hematopoietic stem cell transplantation (HSCT). Methods A retrospective chart review was conducted among SARS-CoV-2-vaccinated HSCT recipients between 2020 and 2022 at a single center in Boston, Massachusetts. Patients age ≥18 years who received doses of Pfizer, Moderna, or J&J vaccines were included. Anti-spike (S) immunoglobulin G (IgG) titer levels were measured using the Roche assay. Responders (≥0.8 U/mL) and nonresponders (<0.8 U/mL) were categorized and analyzed. Multivariable linear and logistic regression were used to estimate the correlation coefficient and odds ratio of response magnitude and status. Results Of 152 HSCT recipients, 141 (92.8%) were responders, with a median (interquartile range [IQR]) anti-S IgG titer of 2500 (107.9-2500) U/mL at a median (IQR) of 80.5 (36-153.5) days from last dose, regardless of the number of doses received. Higher quantitative titers were associated with receipt of more vaccine doses (coeff, 205.79; 95% CI, 30.10 to 381.47; P = .022), being female (coeff, 343.5; 95% CI, -682.6 to -4.4; P = .047), being younger (<65 years; coeff, 365.2; 95% CI, -711.3 to 19.1; P = .039), and not being on anti-CD20 therapy (coeff, -1163.7; 95% CI, -1717.7 to -609.7; P = .001). Being male (odds ratio [OR], 0.11; 95% CI, 0.01 to 0.93; P = .04) and being on anti-CD20 therapy (OR, 0.16; 95% CI, 0.03 to 0.70; P = .016) were associated with nonresponse. Conclusions Overall, most HSCT recipients had high SARS-CoV-2 antibody responses. More vaccine doses improved the magnitude of immune responses. Anti-S IgG monitoring may be useful for identifying attenuated vaccine-induced responses.
Collapse
Affiliation(s)
- Afoke Kokogho
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Trevor A Crowell
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Muneerah Aleissa
- Present affiliation: Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ana-Mihaela Lupan
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Sonya Davey
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jun Bai Park Chang
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Lindsey R Baden
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen R Walsh
- Correspondence: Stephen R. Walsh, MDCM, Division of Infectious Diseases, Brigham & Women’s Hospital, 75 Francis Street, PBB-A-4, Boston, MA 02115 (); or Amy C. Sherman, MD, Division of Infectious Diseases, Brigham & Women’s Hospital, 75 Francis Street, PBB-A-4, Boston, MA 02115 ()
| | - Amy C Sherman
- Correspondence: Stephen R. Walsh, MDCM, Division of Infectious Diseases, Brigham & Women’s Hospital, 75 Francis Street, PBB-A-4, Boston, MA 02115 (); or Amy C. Sherman, MD, Division of Infectious Diseases, Brigham & Women’s Hospital, 75 Francis Street, PBB-A-4, Boston, MA 02115 ()
| |
Collapse
|
23
|
Murase K, Takada K, Arihara Y, Miyanishi K, Kato J. Sotrovimab Treatment for COVID-19 Effective in a B-Cell-Depleted Patient After Anti-CD20 Treatment. Cureus 2023; 15:e41486. [PMID: 37551219 PMCID: PMC10404336 DOI: 10.7759/cureus.41486] [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: 07/06/2023] [Indexed: 08/09/2023] Open
Abstract
Patients with hematologic malignancies are well known to have prolonged COVID-19 pneumonia. The cause has been reported to be B cell depletion after administration of anti-CD20 antibodies. Here, we report a case of COVID-19 pneumonia due to prolonged B-cell depletion after anti-CD20 antibody therapy for malignant lymphoma two years before. Sotrovimab, a neutralizing antibody that was designed to prevent the progression of COVID-19, was successful in preventing the progression to severe disease in this B-cell-depleted patient.
Collapse
Affiliation(s)
- Kazuyuki Murase
- Department of Medical Oncology, Sapporo Medical University, Sapporo, JPN
| | - Kohichi Takada
- Department of Medical Oncology, Sapporo Medical University, Sapporo, JPN
| | - Yohei Arihara
- Department of Medical Oncology, Sapporo Medical University, Sapporo, JPN
| | - Koji Miyanishi
- Department of Medical Oncology, Sapporo Medical University, Sapporo, JPN
| | - Junji Kato
- Department of Medical Oncology, Sapporo Kiyota Hospital, Sapporo, JPN
| |
Collapse
|
24
|
Alfonso-Dunn R, Lin J, Lei J, Liu J, Roche M, De Oliveira A, Raisingani A, Kumar A, Kirschner V, Feuer G, Malin M, Sadiq SA. Humoral and cellular responses to repeated COVID-19 exposure in multiple sclerosis patients receiving B-cell depleting therapies: a single-center, one-year, prospective study. Front Immunol 2023; 14:1194671. [PMID: 37449202 PMCID: PMC10338057 DOI: 10.3389/fimmu.2023.1194671] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/06/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis patients treated with anti-CD20 therapy (aCD20-MS) are considered especially vulnerable to complications from SARS-CoV-2 infection due to severe B-cell depletion with limited viral antigen-specific immunoglobulin production. Therefore, multiple vaccine doses as part of the primary vaccination series and booster updates have been recommended for this group of immunocompromised individuals. Even though much less studied than antibody-mediated humoral responses, T-cell responses play an important role against CoV-2 infection and are induced efficiently in vaccinated aCD20-MS patients. For individuals with such decoupled adaptive immunity, an understanding of the contribution of T-cell mediated immunity is essential to better assess protection against CoV-2 infection. Here, we present results from a prospective, single-center study for the assessment of humoral and cellular immune responses induced in aCD20-MS patients (203 donors/350 samples) compared to a healthy control group (43/146) after initial exposure to CoV-2 spike antigen and subsequent re-challenges. Low rates of seroconversion and RBD-hACE2 blocking activity were observed in aCD20-MS patients, even after multiple exposures (responders after 1st exposure = 17.5%; 2nd exposure = 29.3%). Regarding cellular immunity, an increase in the number of spike-specific monofunctional IFNγ+-, IL-2+-, and polyfunctional IFNγ+/IL-2+-secreting T-cells after 2nd exposure was found most noticeably in healthy controls. Nevertheless, a persistently higher T-cell response was detected in aCD20-MS patients compared to control individuals before and after re-exposure (mean fold increase in spike-specific IFNγ+-, IL-2+-, and IFNγ+/IL-2+-T cells before re-exposure = 3.9X, 3.6X, 3.5X/P< 0.001; after = 3.2X, 1.4X, 2.2X/P = 0.002, P = 0.05, P = 0.004). Moreover, cellular responses against sublineage BA.2 of the currently circulating omicron variant were maintained, to a similar degree, in both groups (15-30% T-cell response drop compared to ancestral). Overall, these results highlight the potential for a severely impaired humoral response in aCD20-MS patients even after multiple exposures, while still generating a strong T-cell response. Evaluating both humoral and cellular responses in vaccinated or infected MS patients on B-cell depletion therapy is essential to better assess individual correlations of immune protection and has implications for the design of future vaccines and healthcare strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Saud A. Sadiq
- Tisch Multiple Sclerosis Research Center of New York, New York, NY, United States
| |
Collapse
|
25
|
Kättström M, Uggla B, Tina E, Kimby E, Norén T, Athlin S. Improved plasmablast response after repeated pneumococcal revaccinations following primary immunization with 13-valent pneumococcal conjugate vaccine or 23-valent pneumococcal polysaccharide vaccine in patients with chronic lymphocytic leukemia. Vaccine 2023; 41:3128-3136. [PMID: 37061372 DOI: 10.1016/j.vaccine.2023.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Patients with chronic lymphocytic leukemia (CLL) show an immune dysfunction with increased risk of infections and poor response to vaccination. Streptococcus pneumoniae is a common cause of morbidity and mortality in CLL patients. In a previous randomized clinical trial, we found a superior immune response in CLL patients receiving conjugated pneumococcal vaccine compared to non-conjugated vaccine. The response to revaccination in CLL patients is scarcely studied. In this study, early humoral response to repeated revaccinations with pneumococcal vaccines was evaluated, by determination of B cell subsets and plasmablast dynamics in peripheral blood. METHOD CLL patients (n = 14) and immunocompetent controls (n = 31) were revaccinated with a 13-valent pneumococcal conjugate vaccine (PCV13) after previous primary immunization (3-6 years ago) with PCV13 or a 23-valent pneumococcal polysaccharide vaccine (PPSV23). Eight weeks after the first revaccination, all CLL patients received a second revaccination with PCV13 or PPSV23. B cell subsets including plasmablasts were analyzed in peripheral blood by flow cytometry, before and after the first and the second revaccination. RESULTS None of the CLL patients, but all controls, had detectable plasmablasts at baseline (p < 0.001). After the first revaccination with PCV13, the plasmablast proportions did not increase in CLL patients (p = 0.13), while increases were seen in controls (p < 0.001). However, after a second revaccination with PCV13 or PPSV23, plasmablasts increased compared to baseline also in CLL patients (p < 0.01). If no response was evident after first revaccination, only a second revaccination with PCV13 increased plasmablasts in contrast to PPSV23 revaccination. Patients with hypogammaglobulinemia and ongoing/previous CLL specific treatment responded poorly, also to a second revaccination. CONCLUSION In CLL patients, pneumococcal revaccination induced minor early plasmablast response compared to controls, but the response improved using a strategy of repeated doses with of conjugated T cell dependent pneumococcal vaccine.
Collapse
Affiliation(s)
- Magdalena Kättström
- Section of Hematology, Department of Medicine, Faculty of Medicine and Health, Örebro University, SE 70185 Örebro, Sweden.
| | - Bertil Uggla
- Section of Hematology, Department of Medicine, Faculty of Medicine and Health, Örebro University, SE 70185 Örebro, Sweden
| | - Elisabet Tina
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Eva Kimby
- Unit of Hematology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Torbjörn Norén
- Department of Laboratory Medicine, Clinical Microbiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Simon Athlin
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, SE 70185 Örebro, Sweden
| |
Collapse
|
26
|
Chevalier K, Genin M, Jean TP, Avouac J, Flipo RM, Georgin-Lavialle S, El Mahou S, Pertuiset E, Pham T, Servettaz A, Marotte H, Domont F, Chazerain P, Devaux M, Mekinian A, Sellam J, Fautrel B, Rouzaud D, Ebstein E, Costedoat-Chalumeau N, Richez C, Hachulla E, Mariette X, Seror R. CovAID: Identification of factors associated with severe COVID-19 in patients with inflammatory rheumatism or autoimmune diseases. Front Med (Lausanne) 2023; 10:1152587. [PMID: 37035330 PMCID: PMC10075312 DOI: 10.3389/fmed.2023.1152587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Autoimmune/inflammatory rheumatic diseases (AIRDs) patients might be at-risk of severe COVID-19. However, whether this is linked to the disease or to its treatment is difficult to determine. This study aimed to identify factors associated with occurrence of severe COVID-19 in AIRD patients and to evaluate whether having an AIRD was associated with increased risk of severe COVID-19 or death. Materials and methods Two databases were analyzed: the EDS (Entrepôt des Données de Santé, Clinical Data Warehouse), including all patients followed in Paris university hospitals and the French multi-center COVID-19 cohort [French rheumatic and musculoskeletal diseases (RMD)]. First, in a combined analysis we compared patients with severe and non-severe COVID-19 to identify factors associated with severity. Then, we performed a propensity matched score case-control study within the EDS database to compare AIRD cases and non-AIRD controls. Results Among 1,213 patients, 195 (16.1%) experienced severe COVID-19. In multivariate analysis, older age, interstitial lung disease (ILD), arterial hypertension, obesity, sarcoidosis, vasculitis, auto-inflammatory diseases, and treatment with corticosteroids or rituximab were associated with increased risk of severe COVID-19. Among 35,741 COVID-19 patients in EDS, 316 having AIRDs were compared to 1,264 Propensity score-matched controls. AIRD patients had a higher risk of severe COVID-19 [aOR = 1.43 (1.08-1.87), p = 0.01] but analysis restricted to rheumatoid arthritis and spondyloarthritis found no increased risk of severe COVID-19 [aOR = 1.11 (0.68-1.81)]. Conclusion In this multicenter study, we confirmed that AIRD patients treated with rituximab or corticosteroids and/or having vasculitis, auto-inflammatory disease, and sarcoidosis had increased risk of severe COVID-19. Also, AIRD patients had, overall, an increased risk of severe COVID-19 compares general population.
Collapse
Affiliation(s)
- Kevin Chevalier
- Department of Rheumatology, Université Paris-Saclay, INSERM UMR 1184: Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Michaël Genin
- University of Lille, CHU Lille, ULR 2694–METRICS: Evaluation des Technologies de Santé et des Pratiques Médicales, Lille, France
| | | | | | | | | | | | | | - Thao Pham
- Hospital Sainte Marguerite, Rheumatology, Marseille, France
| | - Amelie Servettaz
- Hospital Robert Debré, Internal Medicine, Infectious Diseases and Clinical Immunology, Reims, France
| | - Hubert Marotte
- University Hospital of Saint-Étienne, Rheumatology, Saint-Priest-en-Jarez, France
| | - Fanny Domont
- University Hospitals Pitié Salpêtrière - Charles Foix, Internal Medicine and Clinical Immunology, Paris, France
| | - Pascal Chazerain
- Hopital de la Croix Saint-Simon, Rheumatology and Internal Medicine, Paris, France
| | - Mathilde Devaux
- Saint-Germain-en-Laye Intercommunal Hospital Center, Internal Medicine, Poissy, France
| | - Arsene Mekinian
- Hospital Saint-Antoine AP-HP, Internal Medicine, Paris, France
| | - Jérémie Sellam
- Hospital Saint-Antoine AP-HP, Rheumatology, Paris, France
| | - Bruno Fautrel
- Sorbonne Universite – APHP, Pitie Salpetriere Hospital, Department of Rheumatology, Pierre Louis Institute of Epidemiology and Public Health, INSERM UMRS 1136, Paris, France
| | - Diane Rouzaud
- Bichat-Claude Bernard Hospital, Internal Medicine, Paris, France
| | - Esther Ebstein
- Bichat-Claude Bernard Hospital, Rheumatology, Paris, France
| | | | | | - Eric Hachulla
- Department of Internal Medicine and Clinical Immunology, Referral Centre for Centre for Rare Systemic Autoimmune Diseases North and North-West of France (CeRAINO), CHU Lille, University of Lille, INSERM, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Xavier Mariette
- Department of Rheumatology, Université Paris-Saclay, INSERM UMR 1184: Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Raphaèle Seror
- Department of Rheumatology, Université Paris-Saclay, INSERM UMR 1184: Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
27
|
Baroun F, Albasri A, Abutiban F, Alhajeri H. CVAPPS: A Cross-Sectional Study of SARS-CoV-2 Vaccine Acceptance, Perceptions, and Post-Vaccination Side Effects among Rheumatic Disease Patients in Kuwait. Vaccines (Basel) 2023; 11:vaccines11030666. [PMID: 36992250 DOI: 10.3390/vaccines11030666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Purpose: We analyzed data collected for ascertaining severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine acceptance, perceptions, and post-vaccination side effects among Kuwaiti inflammatory rheumatic disease (IRD) patients. Methods: The current cross-sectional study was conducted on patients attending governmental rheumatology clinics across seven hospitals in Kuwait between July and September 2021. We included adults of both sexes who are national/residents of the state of Kuwait and who had a confirmed diagnosis of any IRD disease. Data on patients’ demographics, history of IRD, SARS-CoV-2 infection, vaccination status, as well as post-vaccination side effects and disease flare were collected from the included participants through a self-administered questionnaire. Stata MP/17 for macOS was used for statistical analyses. Results: We included 501 IRD patients, a group with a mean age of 43.38 years and a mean disease duration of 10.46 years. The majority of the included patients were female (79.8%), and the most common primary rheumatology diagnosis was rheumatoid arthritis (42.5%), followed by spondyloarthritis (19.4%) and systemic lupus erythematosus (19.0%). One hundred and five patients (21.0%) had SARS-CoV-2 infection confirmed by PCR-positive swab; of them, 17 patients were hospitalized. None of the included patients were using steroids alone. cDMARDs, bDMARDs, and sDMARDs were reported in 37.3%, 18.0%, and 3.8% of the patients, respectively. Three hundred and fifty-one patients (70.1%) were vaccinated; 40.9% received Pfizer/BioNTech, whereas 28.7% received AstraZeneca/Oxford vaccines. Fear that the vaccination will aggravate the condition or interfere with the present therapy and concerns about its effectiveness as well as its side effects were the most prevalent causes for refusing to accept the SARS-CoV-2 vaccine. Other patients were worried about the paucity of the data because individuals with IRD had been omitted from earlier research, resulting in a dearth of information. The commonly reported post-vaccination side effects were body ache/pain, fatigue, and pain at the injection site (32.1%, 30.3%, and 29.7%, respectively). IRD flare post-SARS-CoV-2 vaccination was self-reported in only 9 patients, and 342 did not report a flare. Conclusions: This study’s findings highlight that SARS-CoV-2 vaccines have an acceptable safety profile, with the majority of their side effects being temporary and mild. The occurrence of flares was low after immunization. Reassurance and trust in the safety of the SARS-CoV-2 vaccination in IRD patients should be reassuring to rheumatologists and vaccine recipients.
Collapse
Affiliation(s)
| | - Anwar Albasri
- Rheumatology Unit, Al-Sabah Hospital, Alsabah, Kuwait
| | | | - Heba Alhajeri
- Rheumatology Unit, Mubarak Al-Kabeer Hospital, Jabriya, Kuwait
| |
Collapse
|
28
|
Peters A, Keating MM, Nikonova A, Doucette S, Prica A. Management of Marginal Zone Lymphoma: A Canadian Perspective. Curr Oncol 2023; 30:1745-1759. [PMID: 36826096 PMCID: PMC9955247 DOI: 10.3390/curroncol30020135] [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: 01/05/2023] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Marginal zone lymphomas (MZL) are a rare, heterogenous group of lymphomas, accounting for 5-17% of indolent non-Hodgkin lymphomas in the western world. They can be further divided into three subtypes: extranodal MZL, splenic MZL, and nodal MZL. These subtypes differ in clinical presentation and behavior, which influences how they are managed. There is currently no standard of care for the treatment of MZL, owing to the difficulty in conducting phase 3 randomized trials in MZL, and the fact that there are limited data on the efficacy of therapy in individual subtypes. Treatment practices are thus largely borrowed from other indolent lymphomas and are based on patient and disease characteristics, as well as access to therapy. This review summarizes the Canadian treatment landscape for MZL and how these therapies may be sequenced in practice.
Collapse
Affiliation(s)
- Anthea Peters
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Correspondence:
| | - Mary-Margaret Keating
- Division of Hematology, Queen Elizabeth II Health Sciences Centre, Dalhousie University, Halifax, NS B3H 2Y9, Canada
| | - Anna Nikonova
- Division of Hematology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | | | - Anca Prica
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| |
Collapse
|
29
|
Passamonti F, Nicastri E, Di Rocco A, Guarini A, Ibatici A, Luminari S, Mikulska M, Visco C. Management of patients with lymphoma and COVID-19: Narrative review and evidence-based practical recommendations. Hematol Oncol 2023; 41:3-15. [PMID: 36251481 PMCID: PMC9874581 DOI: 10.1002/hon.3086] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/29/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023]
Abstract
Patients with hematologic malignancies can be immunocompromized because of their disease, anti-cancer therapy, and concomitant immunosuppressive treatment. Furthermore, these patients are usually older than 60 years and have comorbidities. For all these reasons they are highly vulnerable to infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and have an increased risk of developing severe/critical Coronavirus disease 2019 (COVID-19) compared to the general population. Although COVID-19 vaccination has proven effective in reducing the incidence of severe/critical disease, vaccinated patients with lymphoma may not be protected as they often fail to develop a sufficient antiviral immune response. There is therefore an urgent need to address the management of patients with lymphoma and COVID-19 in the setting of the ongoing pandemic. Passive immunization with monoclonal antibodies against SARS-CoV-2 is a currently available complementary drug strategy to active vaccination for lymphoma patients, while monoclonal antibodies and antiviral drugs (remdesivir, ritonavir-boosted nirmatrelvir, and molnupiravir) have proven effective in preventing the progression to severe/critical COVID-19. In this narrative review we present the most recent data documenting the characteristics and outcomes of patients with concomitant lymphoma and COVID-19. Our ultimate goal is to provide practice-oriented guidance in the management of these vulnerable patients from diagnosis to treatment and follow-up of lymphoma. To this purpose, we will first provide an overview of the main data concerning prognostic factors and fatality rate of lymphoma patients who develop COVID-19; the outcomes of COVID-19 vaccination will also be addressed. We will then discuss current COVID-19 prophylaxis and treatment options for lymphoma patients. Finally, based on the literature and our multidisciplinary experience, we will summarize a set of indications on how to manage patients with lymphoma according to COVID-19 exposure, level of disease severity and former history of infection, as typically encountered in clinical practice.
Collapse
Affiliation(s)
- Francesco Passamonti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.,Hematology, ASST Sette Laghi, Ospedale di Circolo, Varese, Italy
| | - Emanuele Nicastri
- National Institute of Infectious Diseases "L. Spallanzani", IRCCS, Roma, Italy
| | - Alice Di Rocco
- Department of Cellular Biotechnologies and Hematology, Hematology Unit, Sapienza University, Roma, Italy
| | - Attilio Guarini
- Hematology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Adalberto Ibatici
- Hematology Unit and Bone Marrow Transplantation, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Stefano Luminari
- Hematology Unit, Azienda Unità Sanitaria Locale, IRCCS Reggio Emilia, Reggio Emilia, Italy.,Dipartimento CHIMOMO, Università di Modena e Reggio Emilia, Reggio Emilia, Italy
| | - Malgorzata Mikulska
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Division of Infectious Diseases, Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Carlo Visco
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| |
Collapse
|
30
|
Kinzel M, Kalra A, Khanolkar RA, Williamson TS, Li N, Khan F, Puckrin R, Duggan PR, Shafey M, Storek J. Rituximab Toxicity after Preemptive or Therapeutic Administration for Post-Transplant Lymphoproliferative Disorder. Transplant Cell Ther 2023; 29:43.e1-43.e8. [PMID: 36273783 DOI: 10.1016/j.jtct.2022.10.013] [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: 09/07/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Rituximab is commonly used as prevention, preemption, or therapeutically for post-transplant lymphoproliferative disorder (PTLD) after hematopoietic cell transplantation (HCT). Although it is generally assumed that rituximab toxicity (ie, infections resulting from hypogammaglobulinemia and neutropenia) is negligible in relation to mortality due to PTLD, limited evidence supports the validity of this assumption. We sought to determine the impact of rituximab on immunoglobulin levels, neutrophil count, infection density, and mortality outcomes. This study retrospectively analyzed 349 HCT recipients, 289 of whom did not receive rituximab and 60 of whom received rituximab preemptively or therapeutically at a median of 55 days post-transplantation. IgM, IgG, and IgA levels at 6 months and 12 months post-transplantation were lower in patients who received rituximab compared with those who did not (significant at P < .05 for IgM and IgA at 6 months and for IgM and IgG at 12 months). Rituximab recipients also had a higher incidence of severe neutropenia (<.5/nl) between 3 and 24 months (subhazard ratio [SHR], 2.3; P = .020). Regarding non-Epstein-Barr viral infections/PTLD, the rituximab group had a higher infection density between 3 and 24 months compared with the no-rituximab group (3.8 versus 1.6 infections per 365 days at risk; incidence rate ratio, 2.2; P < .001). The rituximab group also had a higher incidence of fatal infections (SHR, 3.1; P = .026), higher nonrelapse mortality (SHR, 2.4; P = .006), and higher overall mortality (hazard ratio, 1.7; P = .033). There were no significant between-group differences in the incidence of clinically significant graft-versus-host disease, graft failure, or relapse. Based on this study, rituximab given for PTLD is associated with substantial morbidity and mortality. Whether the benefit of preemptive rituximab outweighs the risk remains to be determined. © 2022 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.
Collapse
Affiliation(s)
- Megan Kinzel
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Amit Kalra
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rutvij A Khanolkar
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Tyler S Williamson
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Na Li
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Faisal Khan
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Precision Labs, Calgary, Alberta, Canada
| | - Robert Puckrin
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Precision Labs, Calgary, Alberta, Canada
| | - Peter R Duggan
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Precision Labs, Calgary, Alberta, Canada
| | - Mona Shafey
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Precision Labs, Calgary, Alberta, Canada
| | - Jan Storek
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
31
|
Wang KY, Shah P, Skavla B, Fayaaz F, Chi J, Rhodes JM. Vaccination efficacy in patients with chronic lymphocytic leukemia. Leuk Lymphoma 2023; 64:42-56. [PMID: 36270021 DOI: 10.1080/10428194.2022.2133538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is a disorder of mature malignant B cells with multiple elements of immune dysfunction. Infections are common in CLL patients due to complex immunodeficiency. Vaccines are used as preventative measures for common diseases including influenza, pneumococcus, tetanus/diphtheria and shingles in the general population. Vaccines are utilized to mitigate this risk, although there have been some concerns regarding the efficacy of vaccines in the CLL population due to the inherent complex immune dysfunction associated with the disease. In this review, we describe the clinical and laboratory indicators for efficacy of the vaccines in the CLL population (including COVID-19, influenza, pneumonia, herpes zoster, and tetanus) and discuss immunization recommendations for patients with CLL.
Collapse
Affiliation(s)
- Kevin Y Wang
- Department of Internal Medicine, Northwell Health, Manhasset, NY, USA
| | - Pratik Shah
- Department of Internal Medicine, Northwell Health, Manhasset, NY, USA
| | - Brandon Skavla
- Department of Internal Medicine, Northwell Health, Manhasset, NY, USA
| | - Fatima Fayaaz
- Department of Hematology Oncology, Northwell Health, Manhasset, NY, USA
| | - Jeffrey Chi
- Department of Hematology Oncology, Northwell Health, Manhasset, NY, USA
| | - Joanna M Rhodes
- Department of Hematology Oncology, Northwell Health, Manhasset, NY, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
32
|
Liu K, Li J, Xu G. The optimal interval before receiving SARS-COV-2 vaccination for patients who have received Anti-CD 20 monoclonal antibodies. Virulence 2022; 13:2012-2021. [PMID: 36372876 PMCID: PMC9673919 DOI: 10.1080/21505594.2022.2146380] [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: 08/24/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
The optimal interval before receiving SARS-COV-2 vaccination for patients who have received anti-CD 20 monoclonal antibodies remains unclear. We considered original studies up to 29 October 2022 and conducted searches in Embase,Medrxiv, PubMed, and SSRN. We excluded search results that did not match our research question's subject. Human immune response outcomes were analysed inpatients who had previously received anti-CD20 antibody therapy. We analyzed the collected results using sensitivity curves and forest plots. Twenty-eight studies with a total of 1455 subjects receiving anti-CD20 monoclonal antibodies were included in the present analysis. The humoral immune response rates to the time between the last anti-CD20 treatment and vaccination for 3-6 months, 6 months,6-9 months, and 9-12 months were 0.23 (95% CI 0.14 to 0.36), 0.36 (95% CI 0.19 to 0.58), 0.49 (95% CI 0.35 to 0.64) and 0.64 (95% CI 0.48 to 0.77),respectively. The humoral immune response rates were.16 (95% CI 0.03 to 0.57) when B cell was 0/ul, and 0.49 (95% CI 0.38 to 0.61)when B cells were more than 5/ul. The humoral immune response rate for multiple sclerosis was 0.39 (95% CI 0.22 to 0.60) and 0.48 (95% CI 0.29 to 0.68) for B-cell non-Hodgkin lymphoma. The area underneath the curve(AUC) was 0.69 with a cut-off value of 5.5 months. The present results suggested that the optimal interval for SARS-COV-2 vaccination after the final dose of anti-CD20 monoclonal antibody was 5.5 months.
Collapse
Affiliation(s)
- Kexin Liu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Grade 2019, The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Jinyu Li
- Grade 2020, The First Clinical Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
33
|
Sakai A, Morishita T, Suzumura K, Hanatate F, Yoshikawa T, Sasaki N, Lee S, Fujita K, Hara T, Araki H, Tagami A, Murayama M, Yamada R, Iwata A, Sobajima T, Kasahara Y, Matsuzawa Y, Takemura M, Yamamoto Y, Fujigaki H, Saito K, Tsurumi H, Matsunami H. The Trajectory of the COVID-19 Vaccine Antibody Titers Over Time and the Association of Mycophenolate Mofetil in Solid Organ Transplant Recipients. Transplant Proc 2022; 54:2638-2645. [PMID: 36372567 PMCID: PMC9537258 DOI: 10.1016/j.transproceed.2022.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/03/2022] [Accepted: 10/03/2022] [Indexed: 01/07/2023]
Abstract
The COVID-19 vaccine will be safe and effective in solid organ transplant recipients (SOTs). However, the blunted antibody responses were also of concern. Few studies have reported prolonged serologic follow-up after 2 doses of BNT162b2 vaccine in SOTs. We performed a single-center, prospective observational study of 78 SOTs who received 2 doses of BNT162b2 vaccine. We identified the trajectory of antibody titers after vaccination among SOTs with or without mycophenolate mofetil (MMF) or withdrawn from MMF. We found low seroconversion rates (29/42: 69%) and low antibody titers in SOTs treated with MMF. An inverse linear relationship between neutralizing antibody titers and MMF concentration was confirmed in restricted cubic spline plots (P for effect < .01, P for nonlinearity = .08). For the trajectory of antibody responses, seroconversion and improved antibody titers were observed after withdrawal from MMF in SOTs who showed seronegative or low antibody titers at the first visit after 2 doses of vaccine (P for effect < .01, P for nonlinearity < .05, and P for interaction < .01). We identified increased B-cell counts after withdrawal from MMF (P < .01). The recovery of antibody responses was seen in SOTs withdrawn from MMF. The trajectories of antibody responses were modified by MMF administration.
Collapse
Affiliation(s)
- Akiyoshi Sakai
- Department of Clinical Laboratory, Matsunami General Hospital, Gifu, Japan
| | - Tetsuji Morishita
- Department of Internal Medicine, Matsunami General Hospital, Gifu, Japan,Matsunami Research Park, Gifu, Japan,Address correspondence to Tetsuji Morishita, Vice Director of Internal Medicine, Matsunami General Hospital, 185-1 Dendai, Kasamatsu-cho, Hashima-gun, Gifu 501-6062, Japan
| | - Kaori Suzumura
- Department of Clinical Laboratory, Matsunami General Hospital, Gifu, Japan
| | - Fumika Hanatate
- Department of Breast Surgery, Matsunami General Hospital, Gifu, Japan
| | | | | | - Shin Lee
- Department of Hematology, Matsunami General Hospital, Gifu, Japan
| | - Kei Fujita
- Department of Hematology, Matsunami General Hospital, Gifu, Japan
| | - Takeshi Hara
- Department of Hematology, Matsunami General Hospital, Gifu, Japan
| | - Hiroshi Araki
- Department of Gastroenterology, Matsunami General Hospital, Gifu, Japan
| | - Atsushi Tagami
- Department of Gastroenterology, Matsunami General Hospital, Gifu, Japan
| | - Masanori Murayama
- Department of Internal Medicine, Matsunami General Hospital, Gifu, Japan
| | - Rie Yamada
- Department of Internal Medicine, Matsunami General Hospital, Gifu, Japan
| | - Akira Iwata
- Department of Internal Medicine, Matsunami General Hospital, Gifu, Japan
| | - Takuya Sobajima
- Department of Internal Medicine, Matsunami General Hospital, Gifu, Japan
| | - Yukiko Kasahara
- Department of Pediatrics, Matsunami General Hospital, Gifu, Japan
| | - Yoriko Matsuzawa
- Department of Pediatrics, Matsunami General Hospital, Gifu, Japan
| | - Masao Takemura
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Yasuko Yamamoto
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Hidetsugu Fujigaki
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | - Kuniaki Saito
- Department of Advanced Diagnostic System Development, Fujita Health University Graduate School of Health Sciences, Toyoake, Aichi, Japan
| | | | | |
Collapse
|
34
|
Torres M, Corona M, Rodríguez-Mora S, Casado-Fernández G, Zurdo-Castronuño A, Mateos E, Ramos-Martín F, Sánchez-Menéndez C, Murciano-Antón MA, García-Pérez J, Alcamí J, Pérez-Olmeda M, Coiras M, López-Jiménez J, García-Gutiérrez V. Strong Humoral but Not Cellular Immune Responses against SARS-CoV-2 in Individuals with Oncohematological Disease Who Were Treated with Rituximab before Receiving a Vaccine Booster. Cancers (Basel) 2022; 14:5537. [PMID: 36428631 PMCID: PMC9688562 DOI: 10.3390/cancers14225537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
The humoral immune response developed after receiving the full vaccination schedule against COVID-19 is impaired in individuals who received anti-CD20 therapy 6-9 months before vaccination. However, there is little information about the cellular immune responses elicited in these individuals. In this study, we analyzed the humoral and cellular immune responses in 18 individuals with hematological disease who received the last dose of rituximab 13.8 months (IQR 9.4-19) before the booster dose. One month after receiving the booster dose, the seroconversion rate in the rituximab-treated cohort increased from 83.3% to 88.9% and titers of specific IgGs against SARS-CoV-2 increased 1.53-fold (p = 0.0098), while the levels of neutralizing antibodies increased 3.03-fold (p = 0.0381). However, the cytotoxic activity of peripheral blood mononuclear cells (PBMCs) from rituximab-treated individuals remained unchanged, and both antibody-dependent cellular cytotoxicity (ADCC) and direct cellular cytotoxicity (CDD) were reduced 1.7-fold (p = 0.0047) and 2.0-fold (p = 0.0086), respectively, in comparison with healthy donors. Breakthrough infections rate was higher in our cohort of rituximab-treated individuals (33.33%), although most of the infected patients (83.4%) developed a mild form of COVID-19. In conclusion, our findings confirm a benefit in the humoral, but not in the cellular, immune response in rituximab-treated individuals after receiving a booster dose of an mRNA-based vaccine against COVID-19.
Collapse
Affiliation(s)
- Montserrat Torres
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Magdalena Corona
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Sciences, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Sara Rodríguez-Mora
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Guiomar Casado-Fernández
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Faculty of Sciences, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Alejandro Zurdo-Castronuño
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Elena Mateos
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Fernando Ramos-Martín
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Clara Sánchez-Menéndez
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | | | - Javier García-Pérez
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - José Alcamí
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - Mayte Pérez-Olmeda
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Serology Service, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mayte Coiras
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Javier López-Jiménez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Valentín García-Gutiérrez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | | |
Collapse
|
35
|
Krekeler C, Reitnauer L, Bacher U, Khandanpour C, Steger L, Boeckel GR, Klosner J, Tepasse PR, Kemper M, Hennies MT, Mesters R, Stelljes M, Schmitz N, Kerkhoff A, Schliemann C, Mikesch JH, Schmidt N, Lenz G, Bleckmann A, Shumilov E. Efficacy of COVID-19 Booster Vaccines in Patients with Hematologic Malignancies: Experiences in a Real-World Scenario. Cancers (Basel) 2022; 14:5512. [PMID: 36428605 PMCID: PMC9688056 DOI: 10.3390/cancers14225512] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022] Open
Abstract
Background: Two-dose COVID-19 vaccination often results in poor humoral response rates in patients with hematologic malignancies (HMs); yet responses to COVID-19 booster vaccines and the risk of COVID-19 infection post-booster are mostly uncertain. Methods: We included 200 outpatients with HMs and predominantly lymphoid neoplasms (96%, 191/200) in our academic center and reported on the humoral responses, which were assessed by measurement of anti-spike IgG antibodies in peripheral blood as early as 14 days after mRNA-based prime-boost vaccination, as well as factors hampering booster efficacy. Previous basic (double) immunization was applied according to the local recommendations with mRNA- and/or vector-based vaccines. We also report on post-booster COVID-19 breakthrough infections that emerged in the Omicron era and the prophylaxis strategies that were applied to poor and non-responders to booster vaccines. Results: A total of 55% (110/200) of the patients achieved seroconversion (i.e., anti-spike protein IgG antibody titer > 100 AU/mL assessed in median 48 days after prime-boost vaccination) after prime-boost vaccination. Multivariable analyses revealed age, lymphocytopenia, ongoing treatment and prior anti-CD20 B-cell depletion to be independent predictors for booster failure. With each month between anti-CD20-mediated B-cell depletion and booster vaccination, the probability of seroconversion increased by approximately 4% (p < 0.001) and serum−antibody titer (S-AbT) levels increased by 90 AU/mL (p = 0.011). Notably, obinutuzumab treatment was associated with an 85% lower probability for seroconversion after prime-boost vaccination compared to rituximab (p = 0.002). Of poor or non-responders to prime-boost vaccination, 41% (47/114) underwent a second booster and 73% (83/114) underwent passive immunization. COVID-19 breakthrough infections were observed in 15% (29/200) of patients after prime-boost vaccination with predominantly mild courses (93%). Next to seroconversion, passive immunization was associated with a significantly lower risk of COVID-19 breakthrough infections after booster, even in vaccine non-responders (all p < 0.05). In a small proportion of analyzed patients with myeloid neoplasms (9/200), the seroconversion rate was higher compared to those with lymphoid ones (78% vs. 54%, accordingly), while the incidence rate of COVID-19 breakthrough infections was similar (22% vs. 14%, respectively). Following the low frequency of myeloid neoplasms in this study, the results may not be automatically applied to a larger cohort. Conclusions: Patients with HMs are at a high risk of COVID-19 booster vaccine failure; yet COVID-19 breakthrough infections after prime-boost vaccination are predominantly mild. Booster failure can likely be overcome by passive immunization, thereby providing immune protection against COVID-19 and attenuating the severity of COVID-19 courses. Further sophistication of clinical algorithms for preventing post-vaccination COVID-19 breakthrough infections is urgently needed.
Collapse
Affiliation(s)
- Carolin Krekeler
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Lea Reitnauer
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Ulrike Bacher
- Central Hematology Laboratory, Department of Hematology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Cyrus Khandanpour
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
- Department for Hematology and Oncology, University Hospital Schleswig-Holstein, 23564 Luebeck, Germany
| | - Leander Steger
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Göran Ramin Boeckel
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Muenster, Germany
- Department of Medicine D for Nephrology and Rheumatology, University Hospital Münster, 48149 Muenster, Germany
| | - Justine Klosner
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Phil-Robin Tepasse
- Department of Medicine B for Gastroenterology, Hepatology, Endocrinology and Clinical Infectiology, University Hospital Münster, 48149 Muenster, Germany
| | - Marcel Kemper
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Marc Tim Hennies
- Institute of Virology, University Hospital Münster, 48149 Muenster, Germany
| | - Rolf Mesters
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Matthias Stelljes
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Norbert Schmitz
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Andrea Kerkhoff
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Christoph Schliemann
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Nicole Schmidt
- Department of Hematology and Medical Oncology, University Medicine Göttingen (UMG), 37077 Goettingen, Germany
| | - Georg Lenz
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Annalen Bleckmann
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| | - Evgenii Shumilov
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Münster, 48149 Muenster, Germany
| |
Collapse
|
36
|
Totschnig D, Augustin M, Niculescu I, Laferl H, Jansen-Skoupy S, Lehmann C, Wenisch C, Zoufaly A. SARS-CoV-2 Pre-Exposure Prophylaxis with Sotrovimab and Tixagevimab/Cilgavimab in Immunocompromised Patients-A Single-Center Experience. Viruses 2022; 14:2278. [PMID: 36298832 PMCID: PMC9607212 DOI: 10.3390/v14102278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Immunocompromised patients experience reduced vaccine effectiveness and are at higher risk for coronavirus disease 19 (COVID-19) death. Pre-exposure prophylaxis (PrEP) aims to protect these patients. So far, only tixagevimab/cilgavimab is authorized for use as PrEP. This paper aims to provide real-world data on the use of tixagevimab/cilgavimab and sotrovimab as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PrEP in immunocompromised patients, comparing the evolution of antibody levels and reporting the incidence of breakthrough infections. A retrospective, single-center analysis was conducted including 132 immunocompromised patients with inadequate vaccine response, who received COVID-PrEP at our clinic between January and June 2022. Initially, 95 patients received sotrovimab while 37 patients received tixagevimab/cilgavimab. Antibody levels after first PrEP with sotrovimab remain high for several months after infusion (median 10,058 and 7235 BAU/mL after 1 and 3 months, respectively), with higher titers than after tixagevimab/cilgavimab injection even 3 months later (7235 vs. 1647 BAU/mL, p = 0.0007). Overall, breakthrough infections were rare (13/132, 10%) when compared to overall infection rates during this period (over 30% of the Austrian population), with mild disease course and rapid viral clearance (median 10 days). Sotrovimab may be an additional option for SARS-CoV-2 PrEP.
Collapse
Affiliation(s)
- David Totschnig
- Department of Medicine IV, Klinik Favoriten, Vienna Healthcare Group, Kundratstraße 3, 1100 Vienna, Austria
| | - Max Augustin
- Department of Medicine IV, Klinik Favoriten, Vienna Healthcare Group, Kundratstraße 3, 1100 Vienna, Austria
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Iulia Niculescu
- Department of Medicine IV, Klinik Favoriten, Vienna Healthcare Group, Kundratstraße 3, 1100 Vienna, Austria
| | - Hermann Laferl
- Department of Medicine IV, Klinik Favoriten, Vienna Healthcare Group, Kundratstraße 3, 1100 Vienna, Austria
| | - Sonja Jansen-Skoupy
- Department of Laboratory Medicine, Klinik Favoriten, Vienna Healthcare Group, Kundratstraße 3, 1100 Vienna, Austria
| | - Clara Lehmann
- Department I of Internal Medicine, Medical Faculty and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Christoph Wenisch
- Department of Medicine IV, Klinik Favoriten, Vienna Healthcare Group, Kundratstraße 3, 1100 Vienna, Austria
| | - Alexander Zoufaly
- Department of Medicine IV, Klinik Favoriten, Vienna Healthcare Group, Kundratstraße 3, 1100 Vienna, Austria
| |
Collapse
|
37
|
Antibody response to a third booster dose of SARS-CoV-2 vaccination in adults with haematological and solid cancer: a systematic review. Br J Cancer 2022; 127:1827-1836. [PMID: 36224402 PMCID: PMC9555704 DOI: 10.1038/s41416-022-01951-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Patients living with cancer are at a significantly increased risk of morbidity and mortality after infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). This systematic review aims to investigate the current available evidence about the immunogenicity of SARS-CoV-2 booster vaccines in patients living with cancer. METHODS A systematic search was undertaken for studies published until March 1, 2022. A systematic narrative review was undertaken to include all studies that evaluated the efficacy of booster vaccines against SARS-CoV-2 in patients with cancer. RESULTS Fifteen studies encompassing 1205 patients with cancer were included. We found that a booster vaccine dose induced a higher response in patients with solid cancer as compared to haematological malignancies. Recent systemic anticancer therapy does not appear to affect seroconversion in solid organ malignancies, however, there is an association between B-cell depleting therapies and poor seroconversion in haematological patients. CONCLUSIONS Third booster vaccination induces an improved antibody response to SARS-CoV-2 in adults with haematological and solid cancer, relative to patients who only receive two doses. Access to vaccination boosters should be made available to patients at risk of poor immunological responses, and the provision of fourth doses may be of benefit to this vulnerable population. REGISTRATION PROSPERO number CRD42021270420.
Collapse
|
38
|
Shapiro LC, Thakkar A, Gali R, Gonzalez-Lugo JD, Bazarbachi AH, Rahman S, Pradhan K, Fehn K, Abreu M, Kornblum N, Gritsman K, Goldfinger M, Shastri A, Mantzaris I, Braunschweig I, Halmos B, Verma A, McCort M, Bachier-Rodriguez L, Sica RA. High seroconversion rates amongst black and Hispanics with hematologic malignancies after SARS-CoV-2 vaccination. Leuk Lymphoma 2022. [PMID: 35593019 DOI: 10.1101/2021.09.13.21263365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
AbstractIt is well established that COVID-19 carries a higher risk of morbidity and mortality in patients with hematologic malignancies, however, very little data on ethnicity specific responses in this particular patient population currently exist. We established a program of rapid vaccination and evaluation of antibody-mediated response to all EUA COVID-19 vaccines in an inner city minority population to determine the factors that contribute to the poor seroconversion to COVID-19 vaccination in this population. We conducted a cross-sectional cohort study of 126 patients with hematologic malignancies in the outpatient practices of our institution who completed their vaccination series with one of the three FDA EUA COVID-19 vaccines, Moderna, Pfizer, or Johnson & Johnson (J&J). We qualitatively measured Spike IgG production in all patients using the AdviseDx SARS-CoV-2 IgG II assay and quantitatively in 106 patients who completed their vaccination series with at least 14 days after the 2nddose of the Moderna or Pfizer vaccines or 28d after the single J&J vaccine. Patient characteristics were analyzed using standard descriptive statistics and associations between patient characteristics, cancer subtypes, treatments, and vaccine response were assessed using Fisher Exact test or Kruskal-Wallis Rank Sum test. The majority of patients (74%) were minorities. Seventy patients (60%) received Pfizer, 36 patients (31%) Moderna, and 10 patients (9%) J&J. We observed a high-rate of seropositivity (86%) with 16 pts (14%) having a negative Spike IgG. Of the 86 minority patients included, 94% Blacks (30/32) and 87% (39/45) Hispanics showed seropositivity. The factors that contributed to significantly lower seroconversion rates included patients with Non-Hodgkin lymphoma (p=0.005), those who received cytotoxic chemotherapy (p=0.002), IVIG (p=0.01), CAR-T cell therapy (p=0.00002), and CD20 monoclonal antibodies (Ab) (p=0.0000008). Plasma cell neoplasms (p=0.02), immunomodulatory agents (p=0.01), and proteasome inhibitors (p=0.01) had significantly higher seroconversion rates, and those with a history of prior COVID-19 (11%, 12/106) had significantly higher antibody titers (p=0.0003). The positivity rate was 86% (37 seropositive, 6 seronegative) for autologous HSCT and 75% (3 seropositive, 1 seronegative) for allogeneic HSCT. No life-threatening AE were observed. We show high seroconversion rates after SARS-CoV-2 vaccination in non-White patients with hematologic malignancies treated with a wide spectrum of therapeutic modalities. Vaccination is safe, effective, and should be encouraged in most patients with hematologic malignancies. Our minorities based study could be employed as an educational tool to dispel myths and provide data driven evidence to overcome vaccine hesitancy.
Collapse
Affiliation(s)
- Lauren C Shapiro
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Astha Thakkar
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Radhika Gali
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jesus D Gonzalez-Lugo
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abdul-Hamid Bazarbachi
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shafia Rahman
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kith Pradhan
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Karen Fehn
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michelly Abreu
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Noah Kornblum
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kira Gritsman
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mendel Goldfinger
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aditi Shastri
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ioannis Mantzaris
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ira Braunschweig
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amit Verma
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Margaret McCort
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - R Alejandro Sica
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
39
|
Shapiro LC, Thakkar A, Gali R, Gonzalez-Lugo JD, Bazarbachi AH, Rahman S, Pradhan K, Fehn K, Abreu M, Kornblum N, Gritsman K, Goldfinger M, Shastri A, Mantzaris I, Braunschweig I, Halmos B, Verma A, McCort M, Bachier-Rodriguez L, Sica RA. High seroconversion rates amongst black and Hispanics with hematologic malignancies after SARS-CoV-2 vaccination. Leuk Lymphoma 2022; 63:2484-2488. [PMID: 35593019 DOI: 10.1080/10428194.2022.2074988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/25/2022] [Accepted: 05/04/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Lauren C Shapiro
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Astha Thakkar
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Radhika Gali
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jesus D Gonzalez-Lugo
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abdul-Hamid Bazarbachi
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shafia Rahman
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kith Pradhan
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Karen Fehn
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michelly Abreu
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Noah Kornblum
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kira Gritsman
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mendel Goldfinger
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aditi Shastri
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ioannis Mantzaris
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ira Braunschweig
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Amit Verma
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Margaret McCort
- Division of Infectious Diseases, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - R Alejandro Sica
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
40
|
Kister I, Curtin R, Pei J, Perdomo K, Bacon TE, Voloshyna I, Kim J, Tardio E, Velmurugu Y, Nyovanie S, Valeria Calderon A, Dibba F, Stanzin I, Samanovic MI, Raut P, Raposo C, Priest J, Cabatingan M, Winger RC, Mulligan MJ, Patskovsky Y, Silverman GJ, Krogsgaard M. Hybrid and vaccine-induced immunity against SAR-CoV-2 in MS patients on different disease-modifying therapies. Ann Clin Transl Neurol 2022; 9:1643-1659. [PMID: 36165097 PMCID: PMC9538694 DOI: 10.1002/acn3.51664] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE To compare "hybrid immunity" (prior COVID-19 infection plus vaccination) and post-vaccination immunity to SARS CoV-2 in MS patients on different disease-modifying therapies (DMTs) and to assess the impact of vaccine product and race/ethnicity on post-vaccination immune responses. METHODS Consecutive MS patients from NYU MS Care Center (New York, NY), aged 18-60, who completed primary COVID-19 vaccination series ≥6 weeks previously were evaluated for SARS CoV-2-specific antibody responses with electro-chemiluminescence and multiepitope bead-based immunoassays and, in a subset, live virus immunofluorescence-based microneutralization assay. SARS CoV-2-specific cellular responses were assessed with cellular stimulation TruCulture IFNγ and IL-2 assay and, in a subset, with IFNγ and IL-2 ELISpot assays. Multivariate analyses examined associations between immunologic responses and prior COVID-19 infection while controlling for age, sex, DMT at vaccination, time-to-vaccine, and vaccine product. RESULTS Between 6/01/2021 and 11/11/2021, 370 MS patients were recruited (mean age 40.6 years; 76% female; 53% non-White; 22% with prior infection; common DMT classes: ocrelizumab 40%; natalizumab 15%, sphingosine-1-phosphate receptor modulators 13%; and no DMT 8%). Vaccine-to-collection time was 18.7 (±7.7) weeks and 95% of patients received mRNA vaccines. In multivariate analyses, patients with laboratory-confirmed prior COVID-19 infection had significantly increased antibody and cellular post-vaccination responses compared to those without prior infection. Vaccine product and DMT class were independent predictors of antibody and cellular responses, while race/ethnicity was not. INTERPRETATION Prior COVID-19 infection is associated with enhanced antibody and cellular post-vaccine responses independent of DMT class and vaccine type. There were no differences in immune responses across race/ethnic groups.
Collapse
Affiliation(s)
- Ilya Kister
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Ryan Curtin
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Jinglan Pei
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | - Katherine Perdomo
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Tamar E. Bacon
- NYU Multiple Sclerosis Comprehensive Care Center, Department of NeurologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Iryna Voloshyna
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Joseph Kim
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Ethan Tardio
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Yogambigai Velmurugu
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Samantha Nyovanie
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Andrea Valeria Calderon
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Fatoumatta Dibba
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Igda Stanzin
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Marie I. Samanovic
- NYU Langone Vaccine Center, Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Pranil Raut
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | | | | | | | | | - Mark J. Mulligan
- NYU Langone Vaccine Center, Department of MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Yury Patskovsky
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Gregg J. Silverman
- Division of Rheumatology, Department of MedicineNew York University Grossman School of MedicineNew YorkNew York10016USA
| | - Michelle Krogsgaard
- Laura and Isaac Perlmutter Cancer Center and Department of PathologyNew York University Grossman School of MedicineNew YorkNew York10016USA
| |
Collapse
|
41
|
Lueking R, Clark AE, Narasimhan M, Mahimainathan L, Muthukumar A, Larsen CP, SoRelle JA. SARS-CoV-2 coinfections with variant genomic lineages identified by multiplex fragment analysis. Front Genet 2022; 13:942713. [PMID: 36226173 PMCID: PMC9549124 DOI: 10.3389/fgene.2022.942713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Immunocompromised patients can experience prolonged SARS-CoV-2 infections in the setting of a lack of protectivity immunity despite vaccination. As circulating SARS-CoV-2 strains become more heterogeneous, concomitant infection with multiple SARS-CoV-2 variants has become an increasing concern. Immunocompromised patient populations represent potential reservoirs for the emergence of novel SARS-CoV-2 variants through mutagenic change or coinfection followed by recombinatory events. Identification of SARS-CoV-2 coinfections is challenging using traditional next generation sequencing pipelines; however, targeted genotyping approaches can facilitate detection. Here we describe five COVID-19 cases caused by coinfection with different SARS-CoV-2 variants (Delta/Omicron BA.1 and Omicron BA.1/BA.2) as identified by multiplex fragment analysis.
Collapse
Affiliation(s)
- Richard Lueking
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Andrew E. Clark
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Madhusudhanan Narasimhan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Lenin Mahimainathan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Alagarraju Muthukumar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Christian P. Larsen
- Department of Internal Medicine, Division of Infectious Diseases and Geographic Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jeffrey A. SoRelle
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
42
|
Caniza MA, Homsi MR, Bate J, Adrizain R, Ahmed T, Alexander S, Bhattacharyya A, Copado‐Gutierrez JL, Gutierrez I, Lim YY, Morrissey L, Naidu G, Paintsil V, Radhakrishnan N, Mukkada S, Phillips R, Alexander KA, Pritchard‐Jones K. Answers to common questions about COVID-19 vaccines in children with cancer. Pediatr Blood Cancer 2022; 69:e29985. [PMID: 36114651 PMCID: PMC9538403 DOI: 10.1002/pbc.29985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND The SARS-CoV-2 outbreak in 2020 evolved into a global pandemic, and COVID-19 vaccines became rapidly available, including for pediatric patients. However, questions emerged that challenged vaccine acceptance and use. We aimed to answer these questions and give recommendations applicable for use in pediatric patients with cancer by healthcare professionals and the public. METHODS A 12-member global COVID-19 Vaccine in Pediatric Oncology Working Group made up of physicians and nurses from all world regions met weekly from March to July 2021. We used a modified Delphi method to select the top questions. The Working Group, in four-member subgroups, answered assigned questions by providing brief recommendations, followed by a discussion of the rationale for each answer. All Working Group members voted on each recommendation using a scale of 1 to 10, 10 being complete agreement. A "pass" recommendation corresponded to an agreement ≥7.5. RESULTS We selected 15 questions from 173 suggested questions. Based on existing published information, we generated answers for each question as recommendations. The overall average agreement for the 24 recommendations was 9.5 (95% CI 9.4-9.6). CONCLUSION Top COVID-19 vaccine-related questions could be answered using available information. Reports on COVID-19 vaccination and related topics have been published at record speed, aided by available technology and the priority imposed by the pandemic; however, all efforts were made to incorporate emerging information throughout our project. Recommendations will be periodically updated on a dedicated website.
Collapse
Affiliation(s)
- Miguela A. Caniza
- Departments of Global Pediatric Medicine, St. Jude Children's Research HospitalMemphisTennesseeUSA,Departments of Infectious DiseasesSt. Jude Children's Research HospitalMemphisTennesseeUSA,Department of or PediatricsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Maysam R. Homsi
- Departments of Global Pediatric Medicine, St. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Jessica Bate
- Department of Paediatric OncologySouthampton Children's HospitalSouthamptonUK
| | - Riyadi Adrizain
- Department of Child HealthFaculty of Medicine Universitas Padjadjaran Dr. Hasan Sadikin General HospitalBandungIndonesia
| | - Tarek Ahmed
- Department of Pediatric OncologyChildren's Cancer Hospital EgyptCairoEgypt
| | - Sarah Alexander
- Division of Haematology/Oncology, Hospital for Sick Children, Department of PediatricsUniversity of TorontoTorontoCanada
| | | | | | - Ivan Gutierrez
- Department of Pediatric Infectious DiseasesResearch Group Colsubsidio InvestigaClinical Infantil ColsubsidioBogotáColombia,Division of Pediatric Infectious DiseasesClinica Infantil Santa María del LagoBogotáColombia
| | - Yan Yin Lim
- Division of NursingKK Women's and Children's HospitalSingaporeSingapore
| | - Lisa Morrissey
- Department of NursingBoston Children's HospitalBostonMassachusettsUSA
| | - Gita Naidu
- Department of Pediatric OncologyChris Hani Baragwanath Academic HospitalUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Vivian Paintsil
- Department of Child HealthSchool of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Nita Radhakrishnan
- Department of Pediatric Hematology OncologyPost Graduate Institute of Child HealthNoidaIndia
| | - Sheena Mukkada
- Departments of Global Pediatric Medicine, St. Jude Children's Research HospitalMemphisTennesseeUSA,Departments of Infectious DiseasesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Robert Phillips
- Regional Department of Hematology and OncologyLeeds Children's HospitalLeedsUK,Centre for Reviews and DisseminationUniversity of YorkYorkUK
| | - Kenneth A. Alexander
- Department of PediatricsDivision of Infectious DiseasesNemours Children's Hospital‐FloridaOrlandoFloridaUSA
| | - Kathy Pritchard‐Jones
- Developmental Biology and Cancer Research and Teaching DepartmentUCL Great Ormond Street Institute of Child HealthLondonUK
| |
Collapse
|
43
|
Haidar G, Agha M, Bilderback A, Lukanski A, Linstrum K, Troyan R, Rothenberger S, McMahon DK, Crandall MD, Sobolewksi MD, Nathan Enick P, Jacobs JL, Collins K, Klamar-Blain C, Macatangay BJC, Parikh UM, Heaps A, Coughenour L, Schwartz MB, Dueker JM, Silveira FP, Keebler ME, Humar A, Luketich JD, Morrell MR, Pilewski JM, McDyer JF, Pappu B, Ferris RL, Marks SM, Mahon J, Mulvey K, Hariharan S, Updike GM, Brock L, Edwards R, Beigi RH, Kip PL, Wells A, Minnier T, Angus DC, Mellors JW. Prospective Evaluation of Coronavirus Disease 2019 (COVID-19) Vaccine Responses Across a Broad Spectrum of Immunocompromising Conditions: the COVID-19 Vaccination in the Immunocompromised Study (COVICS). Clin Infect Dis 2022; 75:e630-e644. [PMID: 35179197 PMCID: PMC8903515 DOI: 10.1093/cid/ciac103] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND We studied humoral responses after coronavirus disease 2019 (COVID-19) vaccination across varying causes of immunodeficiency. METHODS Prospective study of fully vaccinated immunocompromised adults (solid organ transplant [SOT], hematologic malignancy, solid cancers, autoimmune conditions, human immunodeficiency virus [HIV]) versus nonimmunocompromised healthcare workers (HCWs). The primary outcome was the proportion with a reactive test (seropositive) for immunoglobulin G to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor-binding domain. Secondary outcomes were comparisons of antibody levels and their correlation with pseudovirus neutralization titers. Stepwise logistic regression was used to identify factors associated with seropositivity. RESULTS A total of 1271 participants enrolled: 1099 immunocompromised and 172 HCW. Compared with HCW (92.4% seropositive), seropositivity was lower among participants with SOT (30.7%), hematological malignancies (50.0%), autoimmune conditions (79.1%), solid tumors (78.7%), and HIV (79.8%) (P < .01). Factors associated with poor seropositivity included age, greater immunosuppression, time since vaccination, anti-CD20 monoclonal antibodies, and vaccination with BNT162b2 (Pfizer) or adenovirus vector vaccines versus messenger RNA (mRNA)-1273 (Moderna). mRNA-1273 was associated with higher antibody levels than BNT162b2 or adenovirus vector vaccines after adjusting for time since vaccination, age, and underlying condition. Antibody levels were strongly correlated with pseudovirus neutralization titers (Spearman r = 0.89, P < .0001), but in seropositive participants with intermediate antibody levels, neutralization titers were significantly lower in immunocompromised individuals versus HCW. CONCLUSIONS Antibody responses to COVID-19 vaccines were lowest among SOT and anti-CD20 monoclonal recipients, and recipients of vaccines other than mRNA-1273. Among those with intermediate antibody levels, pseudovirus neutralization titers were lower in immunocompromised patients than HCWs. Additional SARS-CoV-2 preventive approaches are needed for immunocompromised persons, which may need to be tailored to the cause of immunodeficiency.
Collapse
Affiliation(s)
- Ghady Haidar
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mounzer Agha
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Andrew Bilderback
- Wolff Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Amy Lukanski
- Wolff Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Kelsey Linstrum
- Health Care Innovation, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Rachel Troyan
- Wolff Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Scott Rothenberger
- Division of General Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Deborah K McMahon
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Melissa D Crandall
- Clinical Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michele D Sobolewksi
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - P Nathan Enick
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jana L Jacobs
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kevin Collins
- Clinical Analytics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Cynthia Klamar-Blain
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bernard J C Macatangay
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Urvi M Parikh
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy Heaps
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lindsay Coughenour
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Marc B Schwartz
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jeffrey M Dueker
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Fernanda P Silveira
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mary E Keebler
- Department of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Abhinav Humar
- Division of Transplantation, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Matthew R Morrell
- Division of Pulmonary and Critical Care, School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Joseph M Pilewski
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John F McDyer
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bhanu Pappu
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Robert L Ferris
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Stanley M Marks
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - John Mahon
- Clinical Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Katie Mulvey
- Clinical Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sundaram Hariharan
- Division of Transplantation, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Transplant Nephrology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Glenn M Updike
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Magee-Womens Hospital, Pittsburgh, Pennsylvania, USAand
| | - Lorraine Brock
- Wolff Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Robert Edwards
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Magee-Womens Hospital, Pittsburgh, Pennsylvania, USAand
| | - Richard H Beigi
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Magee-Womens Hospital, Pittsburgh, Pennsylvania, USAand
| | - Paula L Kip
- Wolff Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Alan Wells
- Clinical Laboratory, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Tami Minnier
- Wolff Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Derek C Angus
- Health Care Innovation, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - John W Mellors
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
44
|
Steffanoni S, Calimeri T, Laurenge A, Fox CP, Soussain C, Grommes C, Tisi MC, Boot J, Crosbie N, Visco C, Arcaini L, Chaganti S, Sassone MC, Alencar A, Armiento D, Romano I, Dietrich J, Itchaki G, Bruna R, Fracchiolla NS, Arletti L, Venditti A, Booth S, Musto P, Hoang Xuan K, Batchelor T, Cwynarski K, Ferreri AJM. Impact of severe acute respiratory syndrome coronavirus-2 infection on the outcome of primary central nervous system lymphoma treatment: A study of the International PCNSL Collaborative Group. Br J Haematol 2022; 199:507-519. [PMID: 35945164 PMCID: PMC9538907 DOI: 10.1111/bjh.18396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 01/19/2023]
Abstract
To optimise management of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection identifying high-risk patients and maintaining treatment dose intensity is an important issue in patients with aggressive lymphomas. In the present study, we report on the presentation, management, and outcome of an international series of 91 patients with primary central nervous system lymphoma and SARS-CoV-2 infection. SARS-CoV-2 was diagnosed before/during first-line treatment in 64 patients, during follow-up in 21, and during salvage therapy in six. Among the 64 patients infected before/during first-line chemotherapy, 38 (59%) developed pneumonia and 26 (41%) did not clear the virus. Prolonged exposure to steroids before viral infection and/or treatment with high-dose cytarabine favoured pneumonia development and virus persistence and were associated with poorer survival; 81% of patients who did not clear virus died early from coronavirus disease 2019 (COVID-19). Vaccination was associated with lower pneumonia incidence and in-hospital mortality. Chemotherapy was initiated/resumed in 43 (67%) patients, more commonly among patients who did not develop pneumonia, cleared the virus, or did not receive steroids during infection. Chemotherapy resumption in patients with viral persistence should be indicated cautiously as it was associated with a poorer survival (6-month, 70% and 87%, p = 0.07). None of the 21 patients infected during follow-up died from COVID-19, requiring similar measures as infected subjects in the general population.
Collapse
Affiliation(s)
- Sara Steffanoni
- Lymphoma Unit, Department of Onco‐HematologyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Teresa Calimeri
- Lymphoma Unit, Department of Onco‐HematologyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Alice Laurenge
- Service de Neurologie 2‐MazarinHôpitaux Universitaires La Pitié Salpêtrière, APHP, Sorbonne UniversitéParisFrance
| | | | - Carole Soussain
- Hôpital René Huguenin‐Institut Curie, Saint‐CloudParisFrance
| | - Christian Grommes
- Department of NeurologyMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | | | - Jesca Boot
- BarkingHavering and Redbridge University Hospitals NHS TrustLondonUK
| | | | - Carlo Visco
- Department of Medicine, Section of HematologyUniversity of VeronaVeronaItaly
| | - Luca Arcaini
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and Department of Molecular MedicineUniversity of PaviaPaviaItaly
| | | | - Marianna C. Sassone
- Lymphoma Unit, Department of Onco‐HematologyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Alvaro Alencar
- Department of Hematology and OncologyUniversity of Miami/Sylvester Comprehensive Cancer CenterMiamiFloridaUSA
| | | | | | - Jorg Dietrich
- Division of Neuro‐OncologyMassachusetts General Hospital Cancer CenterBostonMassachusettsUSA
| | - Gilad Itchaki
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical CenterPetah‐TikvaIsrael
| | | | | | - Laura Arletti
- Division of HematologyAzienda USL‐IRCCS of Reggio EmiliaReggio EmiliaItaly
| | - Adriano Venditti
- Department of Biomedicine and PreventionUniversity Tor VergataRomeItaly
| | - Stephen Booth
- NIHR Oxford Biomedical Research CentreChurchill HospitalOxfordUK
| | - Pellegrino Musto
- Department of Emergency and Organ Transplantation‘Aldo Moro’ University School of Medicine, and Unit of Hematology and Stem Cell Transplantation, AOUC PoliclinicoBariItaly
| | - Khê Hoang Xuan
- Service de Neurologie 2‐MazarinHôpitaux Universitaires La Pitié Salpêtrière, APHP, Sorbonne UniversitéParisFrance
| | - Tracy T. Batchelor
- Department of NeurologyBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Kate Cwynarski
- Department of HaematologyUniversity College London HospitalLondonUK
| | - Andrés J. M. Ferreri
- Lymphoma Unit, Department of Onco‐HematologyIRCCS San Raffaele Scientific InstituteMilanItaly
| |
Collapse
|
45
|
Pascale SP, Nuccorini R, Pierri T, Di Mare R, Fabio L, Lerose E, Merlino MA, Schiavo P, Amendola A, Brucoli G, Caputo MD, Chitarrelli I, Cimminiello M, Coluzzi S, Filardi NB, Matturro A, Vertone D, Poggiaspalla M, Malaspina F, Musuraca G, Coralluzzo G, Mannarella C, Musto C, Bellettieri AP, Martinelli G, Cerchione C, Pizzuti M. Evaluation of serological response to anti-SARS-CoV-2 mRNA vaccination in hematological patients. Front Immunol 2022; 13:892331. [PMID: 36003404 PMCID: PMC9393554 DOI: 10.3389/fimmu.2022.892331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/07/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction In immunocompromised patients, SARS-CoV-2 mRNA vaccine has been used in Italy from the beginning of the vaccination campaign, but several studies have shown that the serological response of onco-hematological patients was reduced compared to healthy subjects, due to the state of immunosuppression because of both underlying disease and administered therapy. Methods We evaluated the association of anti-SARS-CoV-2 spike IgG titers in 215 hematological patients with clinical and demographic variables to verify if it was possible to identify predictive parameters of serological response, as well as using a control group, consisting of healthy health workers of San Carlo Hospital in Potenza. Anti-SARS-CoV2 IgG titers were evaluated after 30–45 days post second dose vaccine using chemiluminescent microparticle immunoassay technology. Results Patients with hematological malignancies, compared with the control arm, had both a mean concentration of anti-SARS-CoV-2 IgG significantly lower and a seroconversion rate numerically lower. All chronic lymphatic leukemia patients showed levels of antibody titer below the mean concentration, also in only clinical surveillance patients. Comparing serological response in hematological malignancies, only acute leukemia patients who were off therapy had the highest seroconversion rate among the patients’ cohorts and a mean antibody concentration greater than the control arm. Patients treated with steroids and rituximab showed a lower level of anti-SARS-CoV-2 spike IgG. Differences in anti-spike IgG levels among chronic myeloid leukemia patients stratified according to tyrosine kinase inhibitor therapy and molecular response were observed, and they could have interesting implications on the evaluation of the effects of these drugs on the immune system, but having not reached statistical significance at the moment. The cohort of patients who received a stem cell transplant was very heterogeneous because it included different hematological malignancies and different types of transplant; however, a mean concentration of anti-SARS-CoV2 IgG greater than the control arm was reported. Indeed, among patients who performed a transplant for over 6 months only one had a spike IgG concentration below the cutoff. Conclusions Our data confirm reduced serological response in hematological patients after anti-SARS-CoV-2 vaccination. However, we found a great diversity of SARS-CoV-2 antibody response according to types of pathologies and therapies.
Collapse
Affiliation(s)
| | - Roberta Nuccorini
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Teresa Pierri
- UO di Medicina Trasfusionale, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Roberta Di Mare
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Lucia Fabio
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Emilia Lerose
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | | | - Pietro Schiavo
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Angela Amendola
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Gino Brucoli
- UO di Medicina Trasfusionale, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Maria Denise Caputo
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Ida Chitarrelli
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Michele Cimminiello
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Sabrina Coluzzi
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | | | - Angela Matturro
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Domenico Vertone
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Monica Poggiaspalla
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Francesco Malaspina
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Gerardo Musuraca
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Gennaro Coralluzzo
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | - Clara Mannarella
- UOS di Ematologia, Presidio Ospedaliero “Madonna delle Grazie”, Matera, Italy
| | - Clelia Musto
- UO di Medicina Trasfusionale, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
| | | | - Giovanni Martinelli
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- *Correspondence: Claudio Cerchione, ; Michele Pizzuti,
| | - Michele Pizzuti
- UOC di Ematologia, Azienda Ospedaliera Regionale “San Carlo”, Potenza, Italy
- *Correspondence: Claudio Cerchione, ; Michele Pizzuti,
| |
Collapse
|
46
|
Alfonso-Dunn R, Lin J, Kirschner V, Lei J, Feuer G, Malin M, Liu J, Roche M, Sadiq SA. Strong T-cell activation in response to COVID-19 vaccination in multiple sclerosis patients receiving B-cell depleting therapies. Front Immunol 2022; 13:926318. [PMID: 35990701 PMCID: PMC9388928 DOI: 10.3389/fimmu.2022.926318] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
Immunocompromised individuals, including multiple sclerosis (MS) patients on certain immunotherapy treatments, are considered susceptible to complications from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and specific vaccination regimens have been recommended for suitable protection. MS patients receiving anti-CD20 therapy (aCD20-MS) are considered especially vulnerable due to acquired B-cell depletion and impaired antibody production in response to virus infection and COVID-19 vaccination. Here, the humoral and cellular responses are analyzed in a group of aCD20-MS patients (n=43) compared to a healthy control cohort (n=34) during the first 6 months after a 2-dose cycle mRNA-based COVID-19 vaccination. Both IgG antibodies recognizing receptor binding domain (RBD) from CoV-2 spike protein and their blocking activity against RBD-hACE2 binding were significantly reduced in aCD20-MS patients, with a seroconversion rate of only 23.8%. Interestingly, even under conditions of severe B-cell depletion and failed seroconversion, a significantly higher polyfunctional IFNγ+ and IL-2+ T-cell response and strong T-cell proliferation capacity were detected compared to controls. Moreover, no difference in T-cell response was observed between forms of disease (relapsing remitting- vs progressive-MS), anti-CD20 therapy (Rituximab vs Ocrelizumab) and type of mRNA-based vaccine received (mRNA-1273 vs BNT162b2). These results suggest the generation of a partial adaptive immune response to COVID-19 vaccination in B-cell depleted MS individuals driven by a functionally competent T-cell arm. Investigation into the role of the cellular immune response is important to identifying the level of protection against SARS-CoV-2 in aCD20-MS patients and could have potential implications for future vaccine design and application.
Collapse
|
47
|
Ito Y, Honda A, Kurokawa M. COVID-19 mRNA Vaccine in Patients With Lymphoid Malignancy or Anti-CD20 Antibody Therapy: A Systematic Review and Meta-Analysis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e691-e707. [PMID: 35459624 PMCID: PMC8958822 DOI: 10.1016/j.clml.2022.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 04/09/2023]
Abstract
BACKGROUND The humoral response to vaccination in individuals with lymphoid malignancies or those undergoing anti-CD20 antibody therapy is impaired, but details of the response to mRNA vaccines to protect against COVID-19 remain unclear. This systematic review and meta-analysis aimed to characterize the response to COVID-19 mRNA vaccines in patients with lymphoid malignancies or those undergoing anti-CD20 antibody therapy. MATERIALS AND METHODS A literature search retrieved 52 relevant articles, and random-effect models were used to analyze humoral and cellular responses. RESULTS Lymphoid malignancies and anti-CD20 antibody therapy for non-malignancies were significantly associated with lower seropositivity rates (risk ratio 0.60 [95% CI 0.53-0.69]; risk ratio 0.45 [95% CI 0.39-0.52], respectively). Some subtypes (chronic lymphocytic leukemia, treatment-naïve chronic lymphocytic leukemia, myeloma, and non-Hodgkin's lymphoma) exhibited impaired humoral response. Anti-CD20 antibody therapy within 6 months of vaccination decreased humoral response; moreover, therapy > 12 months before vaccination still impaired the humoral response. However, anti-CD20 antibody therapy in non-malignant patients did not attenuate T cell responses. CONCLUSION These data suggest that patients with lymphoid malignancies or those undergoing anti-CD20 antibody therapy experience an impaired humoral response, but cellular response can be detected independent of anti-CD20 antibody therapy. Studies with long-term follow-up of vaccine effectiveness are warranted (PROSPERO registration number: CRD42021265780).
Collapse
Affiliation(s)
- Yusuke Ito
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Akira Honda
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Cell Therapy and Transplantation Medicine, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
48
|
Bellesi S, Sali M, Maiolo E, Pereyra Boza MDC, Alma E, Palucci I, Fatone F, De Maio F, Viscovo M, D'Alò F, De Stefano V, Hohaus S, Sanguinetti M. Anti CD20-based immunochemotherapy abolishes antibody response to Covid-19 mRNA vaccine in lymphoma patients vaccinated during active first line treatment. Leuk Lymphoma 2022; 63:1474-1478. [PMID: 35105258 DOI: 10.1080/10428194.2022.2032042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/14/2022] [Indexed: 10/19/2022]
Affiliation(s)
- Silvia Bellesi
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Michela Sali
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di base, Cliniche intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Maiolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Maria Del Carmen Pereyra Boza
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Eleonora Alma
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Ivana Palucci
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Federica Fatone
- Dipartimento di Scienze Radiologiche ed Ematologiche, Sezione di Ematologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Flavio De Maio
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marcello Viscovo
- Dipartimento di Scienze Radiologiche ed Ematologiche, Sezione di Ematologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco D'Alò
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Dipartimento di Scienze Radiologiche ed Ematologiche, Sezione di Ematologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valerio De Stefano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Dipartimento di Scienze Radiologiche ed Ematologiche, Sezione di Ematologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefan Hohaus
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Dipartimento di Scienze Radiologiche ed Ematologiche, Sezione di Ematologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maurizio Sanguinetti
- Dipartimento Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di base, Cliniche intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
49
|
See KC. Vaccination for the Prevention of Infection among Immunocompromised Patients: A Concise Review of Recent Systematic Reviews. Vaccines (Basel) 2022; 10:800. [PMID: 35632555 PMCID: PMC9144891 DOI: 10.3390/vaccines10050800] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
Vaccination is crucial for avoiding infection-associated morbidity and mortality among immunocompromised patients. However, immunocompromised patients respond less well to vaccinations compared to healthy people, and little is known about the relative efficacy of various vaccines among different immunocompromised states. A total of 54 systematic reviews (22 COVID-19; 32 non-COVID-19) published within the last 5 years in Pubmed® were reviewed. They demonstrated similar patterns within three seroconversion response categories: good (about >60% when compared to healthy controls), intermediate (~40−60%), and poor (about <40%). Good vaccine responses would be expected for patients with chronic kidney disease, human immunodeficiency virus infection (normal CD4 counts), immune-mediated inflammatory diseases, post-splenectomy states, and solid tumors. Intermediate vaccine responses would be expected for patients with anti-cytotoxic T-lymphocyte antigen-4 therapy, hematologic cancer, and human immunodeficiency virus infection (low CD4 counts). Poor vaccine responses would be expected for patients with B-cell-depleting agents (e.g., anti-CD20 therapy), hematopoietic stem-cell transplant, solid organ transplant, and liver cirrhosis. For all vaccine response categories, vaccination should be timed when patients are least immunosuppressed. For the intermediate and poor vaccine response categories, high-dose vaccine, revaccination when patients are less immunosuppressed, checking for seroconversion, additional booster doses, and long-acting monoclonal antibodies may be considered, supplemented by shielding measures.
Collapse
Affiliation(s)
- Kay Choong See
- Division of Respiratory & Critical Care Medicine, Department of Medicine, National University Hospital, Singapore 119228, Singapore
| |
Collapse
|
50
|
[Experts' consensus on severe acute respiratory syndrome coronavirus-2 vaccination in adult patients with hematological diseases in China (2022)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:359-364. [PMID: 35680591 PMCID: PMC9250963 DOI: 10.3760/cma.j.issn.0253-2727.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 11/28/2022]
|