Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 836
Filtrar
1.
Influenza Other Respir Viruses ; 18(3): e13272, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38501337

RESUMO

The emergence of SARS-CoV-2 Omicron variant has led to a complete reconfiguration of the therapeutic landscape, with all monoclonal antibodies having lost any neutralization activity. We report here a case series of 75 immunocompromised patients infected by the Omicron variant who benefited from COVID-19 convalescent plasma (CCP). At Day 28, the overall survival was 76% (95% CI 67-86) with no significant difference in the clinical outcome between patients with hematological malignancies, solid organ transplantation or autoimmune diseases. No safety concern was reported during the course of the study. These results showed that CCP is well tolerated and represents a treatment option for immunocompromised patients who remain highly impacted by the COVID19 epidemic.


Assuntos
COVID-19 , Humanos , COVID-19/terapia , Soroterapia para COVID-19 , SARS-CoV-2 , Imunização Passiva , Hospedeiro Imunocomprometido , Anticorpos Antivirais/uso terapêutico , Anticorpos Neutralizantes
2.
Viruses ; 16(2)2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38399991

RESUMO

Among the anti-Spike monoclonal antibodies (mAbs), the S-309 derivative sotrovimab was the most successful in having the longest temporal window of clinical use, showing a high degree of resiliency to SARS-CoV-2 evolution interrupted only by the appearance of the BA.2.86* variant of interest (VOI). This success undoubtedly reflects rational selection to target a highly conserved epitope in coronavirus Spike proteins. We review here the efficacy of sotrovimab against different SARS-CoV-2 variants in outpatients and inpatients, discussing both randomized controlled trials and real-world evidence. Although it could not be anticipated at the time of its development and introduction, sotrovimab's use in immunocompromised individuals who harbor large populations of variant viruses created the conditions for its eventual demise, as antibody selection and viral evolution led to its eventual withdrawal due to inefficacy against later variant lineages. Despite this, based on observational and real-world data, some authorities have continued to promote the use of sotrovimab, but the lack of binding to newer variants strongly argues for the futility of continued use. The story of sotrovimab highlights the power of modern biomedical science to generate novel therapeutics while also providing a cautionary tale for the need to devise strategies to minimize the emergence of resistance to antibody-based therapeutics.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/uso terapêutico
3.
Clin Immunol ; 260: 109902, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38218210

RESUMO

The devastating impact of COVID-19 on global health shows the need to increase our pandemic preparedness. Recombinant therapeutic antibodies were successfully used to treat and protect at-risk patients from COVID-19. However, the currently circulating Omicron subvariants of SARS-CoV-2 are largely resistant to therapeutic antibodies, and novel approaches to generate broadly neutralizing antibodies are urgently needed. Here, we describe a tetravalent bispecific antibody, A7A9 TVB, which actively neutralized many SARS-CoV-2 variants of concern, including early Omicron subvariants. Interestingly, A7A9 TVB neutralized more variants at lower concentration as compared to the combination of its parental monoclonal antibodies, A7K and A9L. A7A9 also reduced the viral load of authentic Omicron BA.1 virus in infected pseudostratified primary human nasal epithelial cells. Overall, A7A9 displayed the characteristics of a potent broadly neutralizing antibody, which may be suitable for prophylactic and therapeutic applications in the clinics, thus highlighting the usefulness of an effective antibody-designing approach.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Anticorpos Monoclonais/uso terapêutico , Pais , Anticorpos Antivirais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico
4.
Antiviral Res ; 221: 105767, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38040199

RESUMO

Tick-borne encephalitis virus (TBEV), the causative agent of tick-borne encephalitis (TBE), is a medically important flavivirus endemic to the European-Asian continent. Although more than 12,000 clinical cases are reported annually worldwide, there is no anti-TBEV therapy available to treat patients with TBE. Porphyrins are macrocyclic molecules consisting of a planar tetrapyrrolic ring that can coordinate a metal cation. In this study, we investigated the cytotoxicity and anti-TBEV activity of a large series of alkyl- or (het)aryl-substituted porphyrins, metalloporphyrins, and chlorins and characterized their molecular interactions with the viral envelope in detail. Our structure-activity relationship study showed that the tetrapyrrole ring is an essential structural element for anti-TBEV activity, but that the presence of different structurally distinct side chains with different lengths, charges, and rigidity or metal cation coordination can significantly alter the antiviral potency of porphyrin scaffolds. Porphyrins were demonstrated to interact with the TBEV lipid membrane and envelope protein E, disrupt the TBEV envelope and inhibit the TBEV entry/fusion machinery. The crucial mechanism of the anti-TBEV activity of porphyrins is based on photosensitization and the formation of highly reactive singlet oxygen. In addition to blocking viral entry and fusion, porphyrins were also observed to interact with RNA oligonucleotides derived from TBEV genomic RNA, indicating that these compounds could target multiple viral/cellular structures. Furthermore, immunization of mice with porphyrin-inactivated TBEV resulted in the formation of TBEV-neutralizing antibodies and protected the mice from TBEV infection. Porphyrins can thus be used to inactivate TBEV while retaining the immunogenic properties of the virus and could be useful for producing new inactivated TBEV vaccines.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Encefalite Transmitida por Carrapatos , Porfirinas , Humanos , Animais , Camundongos , Vírus da Encefalite Transmitidos por Carrapatos/genética , Anticorpos Antivirais/uso terapêutico , Envelope Viral , Internalização do Vírus , Porfirinas/farmacologia , Porfirinas/uso terapêutico , RNA , Antivirais/farmacologia , Antivirais/uso terapêutico , Cátions/uso terapêutico
6.
Clin Infect Dis ; 78(2): 423-429, 2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-37713176

RESUMO

BACKGROUND: Allogeneic hematopoietic cell transplant recipients (allo-HCTRs) with positive cytomegalovirus (CMV) serology may have false-positive results due to blood product transfusion-associated passive immunity. METHODS: This single-center cohort study included allo-HCTRs with negative baseline (at malignancy diagnosis) CMV serology and indeterminate/low-positive (CMV IgG titer, ≥0.6-<50 U/mL) pretransplant CMV serology with negative pretransplant plasma CMV DNAemia. The CMV status of those patients was reclassified from R+ to R- (CMVR- reclassification group). We compared those patients to allo-HCTRs with negative (CMV IgG titer <0.6 U/mL) pretransplant CMV IgG (CMVR- group). We describe the number and type of patients whose pretransplant CMV status was reclassified from indeterminate/positive to negative. We reviewed all plasma CMV DNAemia tests performed during the first 6 months posttransplant in both groups to assess the safety of this approach. RESULTS: Among 246 (84.5%) of 291 transplanted patients identified as CMVR+ pretransplant, 60 (24.4%) were reclassified from CMV serology indeterminate (N:10)/low-positive (N:50) to R-. Only 1 of 60 patients (1.67%) in the CMVR- reclassification group versus 3 of 44 (6.8%; P = .30) in the CMVR- group developed CMV DNAemia during the follow-up period. There were no significant differences in the number of CMV DNAemia tests performed, CMV DNAemia range, and time posttransplant between the 2 groups. CONCLUSIONS: One of 4 allo-HCT CMVR+ may be falsely flagged as R+, with significant impact on donor selection and prophylaxis administration. A 2-step approach including CMV serology testing at hematologic malignancy diagnosis in allo-HCT candidates and careful review of pretransplant CMV IgG titers may help correctly classify CMV serology status.


Assuntos
Infecções por Citomegalovirus , Transplante de Células-Tronco Hematopoéticas , Humanos , Citomegalovirus , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplantados , Estudos de Coortes , Transplante Homólogo/efeitos adversos , Anticorpos Antivirais/uso terapêutico , Imunoglobulina G , Estudos Retrospectivos
7.
Sci Rep ; 13(1): 21249, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-38040756

RESUMO

The role of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) convalescent plasma in the treatment of Coronavirus Disease 2019 (COVID-19) in immunosuppressed individuals remains controversial. We describe the course of COVID-19 in patients who had received anti-CD20 therapy within the 3 years prior to infection. We compared outcomes between those treated with and those not treated with high titer SARS-CoV2 convalescent plasma. We identified 144 adults treated at Mayo clinic sites who had received anti-CD20 therapies within a median of 5.9 months prior to the COVID-19 index date. About one-third (34.7%) were hospitalized within 14 days and nearly half (47.9%) within 90 days. COVID-19 directed therapy included anti-spike monoclonal antibodies (n = 30, 20.8%), and, among those hospitalized within 14 days (n = 50), remdesivir (n = 45, 90.0%), glucocorticoids (n = 36, 72.0%) and convalescent plasma (n = 24, 48.0%). The duration from receipt of last dose of anti-CD20 therapy did not correlate with outcomes. The overall 90-day mortality rate was 14.7%. Administration of convalescent plasma within 14 days of the COVID-19 diagnosis was not significantly associated with any study outcome. Further study of COVID-19 in CD20-depleted individuals is needed focusing on the early administration of new and potentially combination antiviral agents, associated or not with vaccine-boosted convalescent plasma.


Assuntos
COVID-19 , Adulto , Humanos , COVID-19/terapia , SARS-CoV-2 , RNA Viral , Imunização Passiva , Soroterapia para COVID-19 , Anticorpos Antivirais/uso terapêutico
8.
Front Med ; 17(6): 1117-1134, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38040914

RESUMO

The epidemic of corona virus disease 2019 (COVID-19) caused by severe acute respiratory syndrome Coronavirus 2 and its variants of concern (VOCs) has been ongoing for over 3 years. Antibody therapies encompassing convalescent plasma, hyperimmunoglobulin, and neutralizing monoclonal antibodies (mAbs) applied in passive immunotherapy have yielded positive outcomes and played a crucial role in the early COVID-19 treatment. In this review, the development path, action mechanism, clinical research results, challenges, and safety profile associated with the use of COVID-19 convalescent plasma, hyperimmunoglobulin, and mAbs were summarized. In addition, the prospects of applying antibody therapy against VOCs was assessed, offering insights into the coping strategies for facing new infectious disease outbreaks.


Assuntos
COVID-19 , Doenças Transmissíveis Emergentes , Humanos , Anticorpos Antivirais/uso terapêutico , Doenças Transmissíveis Emergentes/tratamento farmacológico , Tratamento Farmacológico da COVID-19 , COVID-19/terapia , SARS-CoV-2 , Anticorpos Neutralizantes
9.
Int Immunopharmacol ; 125(Pt B): 111159, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37956487

RESUMO

BACKGROUND: Specific antibodies are important for post-vaccination and post-infection immune responses against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The role of antibodies in preventing and treating Coronavirus disease 2019 (COVID-19) in high-risk populations has been highlighted through the use of virus-specific monoclonal antibodies, which has raised the question of immunoglobulin replacement therapy (IRT) used in immunocompromised patients. METHODS: Virus-specific anti-receptor-binding domain (RBD) and anti-nucleocapsid protein (NCAP) antibodies (assessed using a chemiluminescence assay and virus-neutralizing antibodies (virus neutralization test against Delta and Omicron variants)) were analyzed in 20 batches of 10 % (100 mg/mL) immunoglobulin solutions for intravenous IRT from two commercially available producers between January 2022 and March 2023 for clinical use. RESULTS: Anti-RBD and anti-NCAP antibodies were detected in all 20 batches of assessed IRT solutions (mean concentrations of 2817 IU/mL and 2380 IU/mL, respectively). Notably, the concentration of the virus-specific antibodies increased continuously during the follow-up period (from 822.5 IU/mL to 4066.4 IU/mL and 102 IU/mL to 3455.9 IU/mL). These antibodies demonstrated high virus-neutralizing activity against the Delta variant (mean titers of 436 and 325) but were limited to the Omicron variant (mean titers 78 and 70). The differences observed between the two brands were not statistically significant. CONCLUSION: IRT solutions contain high concentrations of anti-SARS-CoV-2 specific antibodies, which may prevent COVID-19; however, the efficacy can be influenced by variable virus-neutralizing activities against different viral strains. Therefore, appropriate IRT should be combined with other approaches, such as vaccination or pre- and post-exposure prophylaxis. Passively transmitted specific antibodies may also lead to false-positive serological test results.


Assuntos
COVID-19 , Humanos , COVID-19/terapia , SARS-CoV-2 , Imunização Passiva , Anticorpos Antivirais/uso terapêutico , Proteínas do Nucleocapsídeo , Receptores Virais , Anticorpos Neutralizantes/uso terapêutico
10.
Antiviral Res ; 220: 105738, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37944822

RESUMO

Coronavirus Disease 2019 (COVID-19) pandemic is severely impacting the world, and tremendous efforts have been made to deal with it. Despite many advances in vaccines and therapeutics, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants remains an intractable challenge. We present a bivalent Receptor Binding Domain (RBD)-specific synthetic antibody, specific for the RBD of wild-type (lineage A), developed from a non-antibody protein scaffold composed of LRR (Leucine-rich repeat) modules through phage display. We further reinforced the unique feature of the synthetic antibody by constructing a tandem dimeric form. The resulting bivalent form showed a broader neutralizing activity against the variants. The in vivo neutralizing efficacy of the bivalent synthetic antibody was confirmed using a human ACE2-expressing mouse model that significantly alleviated viral titer and lung infection. The present approach can be used to develop a synthetic antibody showing a broader neutralizing activity against a multitude of SARS-CoV-2 variants.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Camundongos , Humanos , SARS-CoV-2/genética , Anticorpos , Técnicas de Visualização da Superfície Celular , Glicoproteína da Espícula de Coronavírus/genética , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/uso terapêutico
11.
Int Immunopharmacol ; 124(Pt B): 111020, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37812969

RESUMO

As SARS-CoV-2 variants continue spreading globally, the discovery of broad spectrum therapeutically active antibodies with retaining good protective activity is a global priority. It was reported that infection with SARS-CoV-2 could cause acute lung injury (ALI) in clinical investigations. Therefore, we discovered that anti-RBD scFv is effective against SARS-CoV-2-induced ALI. To begin, we utilized the receptor binding domain (RBD) of spike glycoprotein as a target to produce single-chain antibodies (scFvs) through an intensive phage display technology. The binding affinity and inhibitory effect of the scFvs were evaluated via ELISA and flow cytometry. Moreover, anti-RBD scFv No.35 significantly prevented ALI caused by LPS and SARS-CoV-2 spike RBD protein in mouse model. Thus, the anti-RBD scFv will aid the development of potential antibody treatments and reduce the inflammatory response of SARS-CoV-2.


Assuntos
Lesão Pulmonar Aguda , Anticorpos de Cadeia Única , Animais , Camundongos , Anticorpos Antivirais/uso terapêutico , Ligação Proteica , Anticorpos de Cadeia Única/uso terapêutico , Lesão Pulmonar Aguda/tratamento farmacológico , Anticorpos Neutralizantes/uso terapêutico
12.
J Virol ; 97(11): e0144823, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37855638

RESUMO

IMPORTANCE: The COVID-19 pandemic exposed limitations of conventional antibodies as therapeutics, including high cost, limited potency, ineffectiveness against new viral variants, and primary reliance on injection-only delivery. Nanobodies are single-domain antibodies with therapeutic potentials. We discovered three anti-SARS-CoV-2 nanobodies, named Nanosota-2, -3, and -4, from an immunized alpaca. Nanosota-2 is super potent against prototypic SARS-CoV-2, Nanosota-3 is highly potent against the omicron variant, and Nanosota-4 is effective against both SARS-CoV-1 and SARS-CoV-2. In addition to their super potency and combined broad antiviral spectrum, these nanobodies are cost-effective, can be easily adapted to new viral variants through phage display, and can potentially be administered as inhalers. The Nanosota series are powerful therapeutic candidates to combat circulating SARS-CoV-2 and prepare for possible future coronavirus pandemics.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos de Domínio Único , Humanos , Anticorpos Neutralizantes , Anticorpos Antivirais/uso terapêutico , COVID-19/terapia , Pandemias , Anticorpos de Domínio Único/farmacologia , Glicoproteína da Espícula de Coronavírus
15.
J Virol ; 97(9): e0079023, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37607058

RESUMO

Bats carry genetically diverse severe acute respiratory syndrome-related coronaviruses (SARSr-CoVs). Some of them utilize human angiotensin-converting enzyme 2 (hACE2) as a receptor and cannot efficiently replicate in wild-type mice. Our previous study demonstrated that the bat SARSr-CoV rRsSHC014S induces respiratory infection and lung damage in hACE2 transgenic mice but not wild-type mice. In this study, we generated a mouse-adapted strain of rRsSHC014S, which we named SMA1901, by serial passaging of wild-type virus in BALB/c mice. SMA1901 showed increased infectivity in mouse lungs and induced interstitial lung pneumonia in both young and aged mice after intranasal inoculation. Genome sequencing revealed mutations in not only the spike protein but the whole genome, which may be responsible for the enhanced pathogenicity of SMA1901 in wild-type BALB/c mice. SMA1901 induced age-related mortality similar to that observed in SARS and COVID-19. Drug testing using antibodies and antiviral molecules indicated that this mouse-adapted virus strain can be used to test prophylactic and therapeutic drug candidates against SARSr-CoVs. IMPORTANCE The genetic diversity of SARSr-CoVs in wildlife and their potential risk of cross-species infection highlights the importance of developing a powerful animal model to evaluate the antibodies and antiviral drugs. We acquired the mouse-adapted strain of a bat-origin coronavirus named SMA1901 by natural serial passaging of rRsSHC014S in BALB/c mice. The SMA1901 infection caused interstitial pneumonia and inflammatory immune responses in both young and aged BALB/c mice after intranasal inoculation. Our model exhibited age-related mortality similar to SARS and COVID-19. Therefore, our model will be of high value for investigating the pathogenesis of bat SARSr-CoVs and could serve as a prospective test platform for prophylactic and therapeutic candidates.


Assuntos
Quirópteros , Camundongos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , Animais , Camundongos/virologia , Quirópteros/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Camundongos Endogâmicos BALB C , COVID-19/mortalidade , Síndrome Respiratória Aguda Grave/tratamento farmacológico , Síndrome Respiratória Aguda Grave/mortalidade , Inoculações Seriadas , Antivirais/farmacologia , Antivirais/uso terapêutico , Anticorpos Antivirais/farmacologia , Anticorpos Antivirais/uso terapêutico , Zoonoses Virais/tratamento farmacológico , Zoonoses Virais/transmissão , Zoonoses Virais/virologia , Doenças Pulmonares Intersticiais/tratamento farmacológico , Doenças Pulmonares Intersticiais/virologia , Envelhecimento , Avaliação Pré-Clínica de Medicamentos
16.
Curr Opin Virol ; 61: 101339, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37392670

RESUMO

The filovirus vaccine and the therapeutic monoclonal antibody (mAb) research have made substantial progress. However, existing vaccines and mAbs approved for use in humans are specific to Zaire ebolavirus (EBOV). Since other Ebolavirus species are a continuing threat to public health, the search for broadly protective mAbs has drawn attention. Here, we review viral glycoprotein-targeting mAbs that have proved their broader protective efficacy in animal models. MBP134AF, the most advanced of these new-generation mAb therapies, has recently been deployed in Uganda during the Sudan ebolavirus outbreak. Furthermore, we discuss the measures associated with enhancing antibody therapies and the risks associated with them, including the rise of escape mutations following the mAb treatment and naturally occurring EBOV variants.


Assuntos
Vacinas contra Ebola , Ebolavirus , Doença pelo Vírus Ebola , Animais , Humanos , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/prevenção & controle , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Anticorpos Neutralizantes , Vacinas contra Ebola/uso terapêutico
17.
J Biomed Sci ; 30(1): 59, 2023 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-37525188

RESUMO

BACKGROUND: The COVID-19 pandemic continues to pose a significant worldwide threat to human health, as emerging SARS-CoV-2 Omicron variants exhibit resistance to therapeutic antibodies and the ability to evade vaccination-induced antibodies. Here, we aimed to identify human antibodies (hAbs) from convalescent patients that are potent and broadly neutralizing toward Omicron sublineages. METHODS: Using a single B-cell cloning approach, we isolated BA.5 specific human antibodies. We further examined the neutralizing activities of the most promising neutralizing hAbs toward different variants of concern (VOCs) with pseudotyped virus. RESULTS: Sixteen hAbs showed strong neutralizing activities against Omicron BA.5 with low IC50 values (IC50 < 20 ng/mL). Among four of the most promising neutralizing hAbs (RBD-hAb-B22, -B23, -B25 and -B34), RBD-hAb-B22 exhibited the most potent and broad neutralization profiles across Omicron subvariant pseudoviruses, with low IC50 values (7.7-41.6 ng/mL) and a low PRNT50 value (3.8 ng/mL) in plaque assays with authentic BA.5. It also showed potent therapeutic effects in BA.5-infected K18-hACE2 mice. CONCLUSIONS: Thus, our efficient screening of BA.5-specific neutralizing hAbs from breakthrough infectious convalescent donors successfully yielded hAbs with potent therapeutic potential against multiple SARS-CoV-2 variants.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Animais , Camundongos , Pandemias , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Glicoproteína da Espícula de Coronavírus/genética
18.
Virol Sin ; 38(5): 787-800, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37423308

RESUMO

The weakened protective efficacy of COVID-19 vaccines and antibodies caused by SARS-CoV-2 variants presents a global health emergency, which underscores the urgent need for universal therapeutic antibody intervention for clinical patients. Here, we screened three alpacas-derived nanobodies (Nbs) with neutralizing activity from twenty RBD-specific Nbs. The three Nbs were fused with the Fc domain of human IgG, namely aVHH-11-Fc, aVHH-13-Fc and aVHH-14-Fc, which could specifically bind RBD protein and competitively inhibit the binding of ACE2 receptor to RBD. They effectively neutralized SARS-CoV-2 pseudoviruses D614G, Alpha, Beta, Gamma, Delta, and Omicron sub-lineages BA.1, BA.2, BA.4, and BA.5 and authentic SARS-CoV-2 prototype, Delta, and Omicron BA.1, BA.2 strains. In mice-adapted COVID-19 severe model, intranasal administration of aVHH-11-Fc, aVHH-13-Fc and aVHH-14-Fc effectively protected mice from lethal challenges and reduced viral loads in both the upper and lower respiratory tracts. In the COVID-19 mild model, aVHH-13-Fc, which represents the optimal neutralizing activity among the above three Nbs, effectively protected hamsters from the challenge of SARS-CoV-2 prototype, Delta, Omicron BA.1 and BA.2 by significantly reducing viral replication and pathological alterations in the lungs. In structural modeling of aVHH-13 and RBD, aVHH-13 binds to the receptor-binding motif region of RBD and interacts with some highly conserved epitopes. Taken together, our study illustrated that alpaca-derived Nbs offered a therapeutic countermeasure against SARS-CoV-2, including those Delta and Omicron variants which have evolved into global pandemic strains.


Assuntos
COVID-19 , Camelídeos Americanos , Anticorpos de Domínio Único , Cricetinae , Humanos , Animais , Camundongos , COVID-19/terapia , SARS-CoV-2/genética , Vacinas contra COVID-19 , Anticorpos de Domínio Único/genética , Modelos Animais de Doenças , Imunoglobulina G , Anticorpos Neutralizantes , Anticorpos Antivirais/uso terapêutico , Glicoproteína da Espícula de Coronavírus/genética
19.
Viruses ; 15(7)2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37515166

RESUMO

Chikungunya virus (CHIKV) is a mosquito-transmitted pathogen that causes chikungunya disease (CHIK); the disease is characterized by fever, muscle ache, rash, and arthralgia. This arthralgia can be debilitating and long-lasting, seriously impacting quality of life for years. Currently, there is no specific therapy available for CHIKV infection. We have developed a despeciated equine polyclonal antibody (CHIKV-EIG) treatment against CHIKV and evaluated its protective efficacy in mouse models of CHIKV infection. In immunocompromised (IFNAR-/-) mice infected with CHIKV, daily treatment for five consecutive days with CHIKV-EIG administered at 100 mg/kg starting on the day of infection prevented mortality, reduced viremia, and improved clinical condition as measured by body weight loss. These beneficial effects were seen even when treatment was delayed to 1 day after infection. In immunocompetent mice, CHIKV-EIG treatment reduced virus induced arthritis (including footpad swelling), arthralgia-associated cytokines, viremia, and tissue virus loads in a dose-dependent fashion. Collectively, these results suggest that CHIKV-EIG is effective at preventing CHIK and could be a viable candidate for further development as a treatment for human disease.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Animais , Cavalos , Humanos , Camundongos , Viremia/tratamento farmacológico , Viremia/prevenção & controle , Qualidade de Vida , Vírus Chikungunya/fisiologia , Anticorpos Antivirais/uso terapêutico , Artralgia/tratamento farmacológico , Artralgia/prevenção & controle
20.
Vox Sang ; 118(9): 794-797, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37489640

RESUMO

BACKGROUND AND OBJECTIVES: COVID-19 convalescent plasma (CCP) has retained potency and clinical efficacy against SARS-CoV-2 and is currently of utmost value for seronegative immunocompromised patients. Since most of the effect is due to the vaccine boost of infection-elicited antibodies, there is a theoretical concern that the frequency of suitable donors is declining. MATERIALS AND METHODS: In this single-institution serosurvey, we screened 599 consecutive donors attending our area in two different seasons (300 in November 2022 and 299 in February 2023) using the Abbott Alinity® anti-Spike immunoglobulin G assay. RESULTS: More than 80% of random donors qualify according to the FDA criteria for high-titre CCP (>4350 AU/mL), with a stable trend. CONCLUSION: Despite reduced anti-Spike vaccine boost deployment in the general population, we have shown here that high-titre CCP units are easier than ever to procure. This finding also has implications for the derivation of standard immunoglobulins, which are finally approaching the potency of hyperimmune serum and could soon represent an alternative to CCP.


Assuntos
COVID-19 , Vacinas Anticâncer , Humanos , Doadores de Sangue , COVID-19/terapia , Soroterapia para COVID-19 , SARS-CoV-2 , Itália , Imunoglobulina G , Anticorpos Antivirais/uso terapêutico , Imunização Passiva , Anticorpos Neutralizantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...