Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nat Biomed Eng ; 8(4): 361-379, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38486104

RESUMO

Mice adoptively transferred with mouse B cells edited via CRISPR to express human antibody variable chains could help evaluate candidate vaccines and develop better antibody therapies. However, current editing strategies disrupt the heavy-chain locus, resulting in inefficient somatic hypermutation without functional affinity maturation. Here we show that these key B-cell functions can be preserved by directly and simultaneously replacing recombined mouse heavy and kappa chains with those of human antibodies, using a single Cas12a-mediated cut at each locus and 5' homology arms complementary to distal V segments. Cells edited in this way to express the human immunodeficiency virus type 1 (HIV-1) broadly neutralizing antibody 10-1074 or VRC26.25-y robustly hypermutated and generated potent neutralizing plasma in vaccinated mice. The 10-1074 variants isolated from the mice neutralized a global panel of HIV-1 isolates more efficiently than wild-type 10-1074 while maintaining its low polyreactivity and long half-life. We also used the approach to improve the potency of anti-SARS-CoV-2 antibodies against recent Omicron strains. In vivo affinity maturation of B cells edited at their native loci may facilitate the development of broad, potent and bioavailable antibodies.


Assuntos
Anticorpos Neutralizantes , Linfócitos B , COVID-19 , Anticorpos Anti-HIV , HIV-1 , SARS-CoV-2 , Animais , Humanos , Camundongos , Linfócitos B/imunologia , HIV-1/imunologia , SARS-CoV-2/imunologia , Anticorpos Anti-HIV/imunologia , Anticorpos Neutralizantes/imunologia , COVID-19/imunologia , COVID-19/virologia , Afinidade de Anticorpos/imunologia , Sistemas CRISPR-Cas/genética , Vacinas contra COVID-19/imunologia , Anticorpos Antivirais/imunologia , Camundongos Endogâmicos C57BL
3.
bioRxiv ; 2023 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-37961481

RESUMO

CRISPR-edited murine B cells engineered to express human antibody variable chains proliferate, class switch, and secrete these antibodies in vaccinated mice. However, current strategies disrupt the heavy-chain locus, resulting in inefficient somatic hypermutation without functional affinity maturation. Here we show that recombined murine heavy- and kappa-variable genes can be directly and simultaneously overwritten, using Cas12a-mediated cuts at their 3'-most J segments and 5' homology arms complementary to distal V segments. Cells edited in this way to express the HIV-1 broadly neutralizing antibodies 10-1074 or VRC26.25-y robustly hypermutated and generated potent neutralizing plasma in vaccinated recipient mice. 10-1074 variants isolated from these mice bound and neutralized HIV-1 envelope glycoprotein more efficiently than wild-type 10-1074 while maintaining or improving its already low polyreactivity and long in vivo half-life. We further validated this approach by generating substantially broader and more potent variants of the anti-SARS-CoV-2 antibodies ZCB11 and S309. Thus, B cells edited at their native loci affinity mature, facilitating development of broad, potent, and bioavailable antibodies and expanding the potential applications of engineered B cells.

4.
Respir Res ; 24(1): 204, 2023 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-37598171

RESUMO

BACKGROUND: Pulmonary hypertension (PH) is a lethal vascular disease with limited therapeutic options. The mechanistic connections between alveolar hypoxia and PH are not well understood. The aim of this study was to investigate the role of mitotic regulator Polo-like kinase 1 (PLK1) in PH development. METHODS: Mouse lungs along with human pulmonary arterial smooth muscle cells and endothelial cells were used to investigate the effects of hypoxia on PLK1. Hypoxia- or Sugen5416/hypoxia was applied to induce PH in mice. Plk1 heterozygous knockout mice and PLK1 inhibitors (BI 2536 and BI 6727)-treated mice were checked for the significance of PLK1 in the development of PH. RESULTS: Hypoxia stimulated PLK1 expression through induction of HIF1α and RELA. Mice with heterozygous deletion of Plk1 were partially resistant to hypoxia-induced PH. PLK1 inhibitors ameliorated PH in mice. CONCLUSIONS: Augmented PLK1 is essential for the development of PH and is a druggable target for PH.


Assuntos
Hipertensão Pulmonar , Humanos , Animais , Camundongos , Hipertensão Pulmonar/genética , Células Endoteliais , Proteínas de Ciclo Celular/genética , Hipóxia , Camundongos Knockout , Quinase 1 Polo-Like
5.
Antib Ther ; 6(3): 157-169, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37492588

RESUMO

In vitro display technologies have been successfully utilized for the discovery and evolution of monoclonal antibodies (mAbs) for diagnostic and therapeutic applications, with phage display and yeast display being the most commonly used platforms due to their simplicity and high efficiency. As their prokaryotic or lower eukaryotic host organisms typically have no or different post-translational modifications, several mammalian cell-based display and screening technologies for isolation and optimization of mAbs have emerged and are being developed. We report here a novel and useful mammalian cell display platform based on the PiggyBac transposon system to display mAbs in a single-chain Fab (scFab) format on the surface of HEK293F cells. Immune rabbit antibody libraries encompassing ~7 × 107 independent clones were generated in an all-in-one transposon vector, stably delivered into HEK293F cells and displayed as an scFab with rabbit variable and human constant domains. After one round of magnetic activated cell sorting and two rounds of fluorescence activated cell sorting, mAbs with high affinity in the subnanomolar range and cross-reactivity to the corresponding human and mouse antigens were identified, demonstrating the power of this platform for antibody discovery. We developed a highly efficient mammalian cell display platform based on the PiggyBac transposon system for antibody discovery, which could be further utilized for humanization as well as affinity and specificity maturation.

6.
Future Oncol ; 19(23): 1613-1626, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37377070

RESUMO

Background: We explored whether a model based on contrast-enhanced computed tomography radiomics features and clinicopathological factors can evaluate preoperative lymphovascular invasion (LVI) in patients with gastric cancer (GC) with Lauren classification. Methods: Based on clinical and radiomic characteristics, we established three models: Clinical + Arterial phase_Radcore, Clinical + Venous phase_Radcore and a combined model. The relationship between Lauren classification and LVI was analyzed using a histogram. Results: We retrospectively analyzed 495 patients with GC. The areas under the curve of the combined model were 0.8629 and 0.8343 in the training and testing datasets, respectively. The combined model showed a superior performance to the other models. Conclusion: CECT-based radiomics models can effectively predict preoperative LVI in GC patients with Lauren classification.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/diagnóstico por imagem , Neoplasias Gástricas/cirurgia , Neoplasias Gástricas/patologia , Estudos Retrospectivos , Tomografia Computadorizada por Raios X/métodos , Metástase Linfática , Meios de Contraste
7.
J Mol Biol ; 435(10): 168085, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37019174

RESUMO

Monoclonal antibody (mAb)-based biologics are well established treatments of cancer. Antibody discovery campaigns are typically directed at a single target of interest, which inherently limits the possibility of uncovering novel antibody specificities or functionalities. Here, we present a target-unbiased approach for antibody discovery that relies on generating mAbs against native target cell surfaces via phage display. This method combines a previously reported method for improved whole-cell phage display selections with next-generation sequencing analysis to efficiently identify mAbs with the desired target cell reactivity. Applying this method to multiple myeloma cells yielded a panel of >50 mAbs with unique sequences and diverse reactivities. To uncover the identities of the cognate antigens recognized by this panel, representative mAbs from each unique reactivity cluster were used in a multi-omic target deconvolution approach. From this, we identified and validated three cell surface antigens: PTPRG, ICAM1, and CADM1. PTPRG and CADM1 remain largely unstudied in the context of multiple myeloma, which could warrant further investigation into their potential as therapeutic targets. These results highlight the utility of optimized whole-cell phage display selection methods and could motivate further interest in target-unbiased antibody discovery workflows.


Assuntos
Anticorpos Monoclonais , Antígenos , Biblioteca de Peptídeos , Humanos , Especificidade de Anticorpos , Molécula 1 de Adesão Celular , Técnicas de Visualização da Superfície Celular/métodos , Multiômica , Mieloma Múltiplo/genética
8.
J Immunother Cancer ; 10(11)2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36442911

RESUMO

BACKGROUND: Despite numerous therapeutic options, safe and curative therapy is unavailable for most patients with chronic lymphocytic leukemia (CLL). A drawback of current therapies such as the anti-CD20 monoclonal antibody (mAb) rituximab is the elimination of all healthy B cells, resulting in impaired humoral immunity. We previously reported the identification of a patient-derived, CLL-binding mAb, JML-1, and identified sialic acid-binding immunoglobulin-like lectin-6 (Siglec-6) as the target of JML-1. Although little is known about Siglec-6, it appears to be an attractive target for cancer immunotherapy due to its absence on most healthy cells and tissues. METHODS: We used a target-specific approach to mine for additional patient-derived anti-Siglec-6 mAbs. To assess the therapeutic utility of targeting Siglec-6 in the context of CLL, T cell-recruiting bispecific antibodies (T-biAbs) that bind to Siglec-6 and CD3 were engineered into single-chain variable fragment-Fc and dual-affinity retargeting (DART)-Fc constructs. T-biAbs were evaluated for their activity in vitro, ex vivo, and in vivo. RESULTS: We discovered the anti-Siglec-6 mAbs RC-1 and RC-2, which bind with higher affinity than JML-1 yet maintain similar specificity. Both JML-1 and RC-1 T-biAbs were effective at activating T cells and killing Siglec-6+ target cells. The RC-1 clone in the DART-Fc format was the most potent T-biAb tested and was the only anti-Siglec-6 T-biAb that eliminated Siglec-6+ primary CLL cells via autologous T cells at pathological T-to-CLL cell ratios. Tested at healthy T-to-B cell ratios, it also eliminated a Siglec-6+ fraction of primary B cells from healthy donors. The subpicomolar potency of the DART-Fc format was attributed to the reduction in the length and flexibility of the cytolytic synapse. Furthermore, the RC-1 T-biAb was effective at clearing MEC1 CLL cells in vivo and demonstrated a circulatory half-life of over 7 days. CONCLUSION: Siglec-6-targeting T-biAbs are highly potent and specific for eliminating Siglec-6+ leukemic and healthy B cells while sparing Siglec-6- healthy B cells, suggesting a unique treatment strategy for CLL with diminished suppression of humoral immunity. Our data corroborate reports that T-biAb efficacy is dependent on synapse geometry and reveal that synapse architecture can be tuned via antibody engineering. Our fully human anti-Siglec-6 antibodies and T-biAbs have potential for cancer immunotherapy. TRIAL REGISTRATION NUMBER: NCT00923507.


Assuntos
Leucemia Linfocítica Crônica de Células B , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Linfócitos T , Linfócitos B , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Imunoterapia
9.
Oncogene ; 41(34): 4104-4114, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35859167

RESUMO

The success of chimeric antigen receptor T cell (CAR-T) therapy in the treatment of hematologic malignancies has prompted the development of numerous CAR-T technologies, including switchable CAR-T (sCAR-T) systems that combine a universal CAR-T with bispecific adapter proteins. Owing to their controllability and versatility, sCAR-Ts have received considerable attention. To explore the therapeutic utility of sCAR-Ts targeting the receptor tyrosine kinase ROR1, which is expressed in hematologic and solid malignancies, and to identify bispecific adaptor proteins that efficiently mediate universal CAR-T engagement, a panel of switches based on ROR1-targeting Fabs with different epitopes and affinities was compared in in vitro and in vivo models of ROR1-expressing cancers. For switches targeting overlapping or identical epitopes, potency correlated with affinity. Surprisingly, however, we identified a switch targeting a unique epitope with low affinity but mediating potent and selective antitumor activity in vitro and in vivo. Converted to a conventional CAR-T, the same anti-ROR1 mAb (324) outperformed a clinically investigated conventional CAR-T that is based on an anti-ROR1 mAb (R12) with ~200-fold higher affinity. Thus, demonstrating therapeutic utility on their own, sCAR-Ts also facilitate higher throughput screening for the identification of conventional CAR-T candidates for preclinical and clinical studies.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Linhagem Celular Tumoral , Epitopos , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo
10.
Front Oncol ; 12: 784839, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35311124

RESUMO

Purpose: This study aimed to assess the diagnostic performance and the added value to radiologists of different levels of a computer-aided diagnosis (CAD) system for the detection of pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) in patients with breast cancer. Besides, to investigate whether tumor molecular typing is associated with the efficiency of diagnosis of the CAD systems. Methods: 470 patients were identified with breast cancers who underwent NAC and post MR imaging between January 2016 and March 2019. The diagnostic performance of radiologists of different levels and the CAD system were compared. The added value of the CAD system was assessed and subgroup analyses were performed according to the tumor molecular typing. Results: Among 470 patients, 123 (26%) underwent pCR. The CAD system showed a comparable specificity as the senior radiologist (83.29% vs. 84.15%, p=0.488) and comparable area under the curve (AUC) (0.839 vs. 0.835, p =0.452). The performance of all radiologists significantly improved when aided by the CAD system (P<0.05), And there were no statistical differences in terms of sensitivity, specificity and accuracy between the two groups with CAD assistance(p>0.05).The AUC values for identifying pCR in TN patients were significant (0.883, 95%CI: 0.801-0.964, p < 0.001). Conclusion: The CAD system assessed in this study improves the performance of all radiologists, regardless of experience. The molecular typing of breast cancer is potential influencer of CAD diagnostic performance.

11.
Future Oncol ; 18(8): 991-1001, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34894719

RESUMO

Background: To determine suitable optimal classifiers and examine the general applicability of computer-aided classification to compare the differences between a computer-aided system and radiologists in predicting pathological complete response (pCR) from patients with breast cancer receiving neoadjuvant chemotherapy. Methods: We analyzed a total of 455 masses and used the U-Net network and ResNet to execute MRI segmentation and pCR classification. The diagnostic performance of radiologists, the computer-aided system and a combination of radiologists and computer-aided system were compared using receiver operating characteristic curve analysis. Results: The combination of radiologists and computer-aided system had the best performance for predicting pCR with an area under the curve (AUC) value of 0.899, significantly higher than that of radiologists alone (AUC: 0.700) and computer-aided system alone (AUC: 0.835). Conclusion: An automated classification system is feasible to predict the pCR to neoadjuvant chemotherapy in patients with breast cancer and can complement MRI.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Terapia Neoadjuvante , Radiologistas , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Curva ROC , Estudos Retrospectivos
12.
Antib Ther ; 4(4): 222-227, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34805745

RESUMO

Antibody-drug conjugates (ADCs) are targeted therapeutics generated by conjugation of cytotoxic small molecules to monoclonal antibodies (mAbs) via chemical linkers. Due to their selective delivery of toxic payloads to antigen-positive cancer cells, ADCs demonstrate wider therapeutic indexes compared with conventional chemotherapy. After decades of intensive research and development, significant advances have been made in the field, leading to a total of 10 U.S. food and drug administration (FDA)-approved ADCs to treat cancer patients. Currently, ~80 ADCs targeting different antigens are under clinical evaluation for treatment of either hematological or solid malignancies. Notably, three ADCs targeting the same oncofetal protein, receptor tyrosine kinase like orphan receptor 1 (ROR1), have attracted considerable attention when they were acquired or licensed successively in the fourth quarter of 2020 by three major pharmaceutical companies. Apparently, ROR1 has emerged as an attractive target for cancer therapy. Since all the components of ADCs, including the antibody, linker and payload, as well as the conjugation method, play critical roles in ADC's efficacy and performance, their choice and combination will determine how far they can be advanced. This review summarizes the design and development of current anti-ROR1 ADCs and highlights an emerging trend to target ROR1 for cancer therapy.

13.
Blood Adv ; 5(16): 3152-3162, 2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34424320

RESUMO

Antibody-drug conjugates directed against tumor-specific targets have allowed targeted delivery of highly potent chemotherapy to malignant cells while sparing normal cells. Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncofetal protein with limited expression on normal adult tissues and is overexpressed on the surface of malignant cells in mantle cell lymphoma, acute lymphocytic leukemia with t(1;19)(q23;p13) translocation, and chronic lymphocytic leukemia. This differential expression makes ROR1 an attractive target for antibody-drug conjugate therapy, especially in malignancies such as mantle cell lymphoma and acute lymphocytic leukemia, in which systemic chemotherapy remains the gold standard. Several preclinical and phase 1 clinical studies have established the safety and effectiveness of anti-ROR1 monoclonal antibody-based therapies. Herein we describe a humanized, first-in-class anti-ROR1 antibody-drug conjugate, huXBR1-402-G5-PNU, which links a novel anti-ROR1 antibody (huXBR1-402) to a highly potent anthracycline derivative (PNU). We found that huXBR1-402-G5-PNU is cytotoxic to proliferating ROR1+ malignant cells in vitro and suppressed leukemia proliferation and extended survival in multiple models of mice engrafted with human ROR1+ leukemia. Lastly, we show that the B-cell lymphoma 2 (BCL2)-dependent cytotoxicity of huXBR1-402-G5-PNU can be leveraged by combined treatment strategies with the BCL2 inhibitor venetoclax. Together, our data present compelling preclinical evidence for the efficacy of huXBR1-402-G5-PNU in treating ROR1+ hematologic malignancies.


Assuntos
Neoplasias Hematológicas , Imunoconjugados , Leucemia Linfocítica Crônica de Células B , Linfoma de Célula do Manto , Animais , Anticorpos Monoclonais , Linfoma de Célula do Manto/tratamento farmacológico , Linfoma de Célula do Manto/genética , Camundongos
14.
Front Oncol ; 11: 693339, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34249745

RESUMO

BACKGROUND: There is a demand for additional alternative methods that can allow the differentiation of the breast tumor into molecular subtypes precisely and conveniently. PURPOSE: The present study aimed to determine suitable optimal classifiers and investigate the general applicability of computer-aided diagnosis (CAD) to associate between the breast cancer molecular subtype and the extracted MR imaging features. METHODS: We analyzed a total of 264 patients (mean age: 47.9 ± 9.7 years; range: 19-81 years) with 264 masses (mean size: 28.6 ± 15.86 mm; range: 5-91 mm) using a Unet model and Gradient Tree Boosting for segmentation and classification. RESULTS: The tumors were segmented clearly by the Unet model automatically. All the extracted features which including the shape features,the texture features of the tumors and the clinical features were input into the classifiers for classification, and the results showed that the GTB classifier is superior to other classifiers, which achieved F1-Score 0.72, AUC 0.81 and score 0.71. Analyzed the different features combinations, we founded that the texture features associated with the clinical features are the optimal features to different the breast cancer subtypes. CONCLUSION: CAD is feasible to differentiate the breast cancer subtypes, automatical segmentation were feasible by Unet model and the extracted texture features from breast MR imaging with the clinical features can be used to help differentiating the molecular subtype. Moreover, in the clinical features, BPE and age characteristics have the best potential for subtype.

15.
PLoS Pathog ; 17(4): e1009501, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33836016

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein mediates infection of cells expressing angiotensin-converting enzyme 2 (ACE2). ACE2 is also the viral receptor of SARS-CoV (SARS-CoV-1), a related coronavirus that emerged in 2002-2003. Horseshoe bats (genus Rhinolophus) are presumed to be the original reservoir of both viruses, and a SARS-like coronavirus, RaTG13, closely related to SARS-CoV-2, has been identified in one horseshoe-bat species. Here we characterize the ability of the S-protein receptor-binding domains (RBDs) of SARS-CoV-1, SARS-CoV-2, pangolin coronavirus (PgCoV), RaTG13, and LyRa11, a bat virus similar to SARS-CoV-1, to bind a range of ACE2 orthologs. We observed that the PgCoV RBD bound human ACE2 at least as efficiently as the SARS-CoV-2 RBD, and that both RBDs bound pangolin ACE2 efficiently. We also observed a high level of variability in binding to closely related horseshoe-bat ACE2 orthologs consistent with the heterogeneity of their RBD-binding regions. However five consensus horseshoe-bat ACE2 residues enhanced ACE2 binding to the SARS-CoV-2 RBD and neutralization of SARS-CoV-2 pseudoviruses by an enzymatically inactive immunoadhesin form of human ACE2 (hACE2-NN-Fc). Two of these mutations impaired neutralization of SARS-CoV-1 pseudoviruses. An hACE2-NN-Fc variant bearing all five mutations neutralized both SARS-CoV-2 pseudovirus and infectious virus more efficiently than wild-type hACE2-NN-Fc. These data suggest that SARS-CoV-1 and -2 originate from distinct bat species, and identify a more potently neutralizing form of soluble ACE2.


Assuntos
Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/imunologia , COVID-19/imunologia , COVID-19/virologia , Quirópteros/metabolismo , SARS-CoV-2/genética , Animais , COVID-19/genética , Quirópteros/genética , Especificidade de Hospedeiro/genética , Especificidade de Hospedeiro/imunologia , Humanos , Modelos Moleculares , Mutação , Ligação Proteica/genética , Ligação Proteica/fisiologia , Receptores Virais/metabolismo , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo
16.
Mol Ther Oncolytics ; 20: 387-398, 2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33614919

RESUMO

Metastatic medullary thyroid cancer (MTC) is a rare but often aggressive thyroid malignancy with a 5-year survival rate of less than 40% and few effective therapeutic options. Adoptive T cell immunotherapy using chimeric antigen receptor (CAR)-modified T cells (CAR Ts) is showing encouraging results in the treatment of cancer, but development is challenged by the availability of suitable target antigens. We identified glial-derived neurotrophic factor (GDNF) family receptor alpha 4 (GFRα4) as a putative antigen target for CAR-based therapy of MTC. We show that GFRα4 is highly expressed in MTC, in parafollicular cells within the thyroid from which MTC originates, and in normal thymus. We isolated two single-chain variable fragments (scFvs) targeting GFRα4 isoforms a and b by antibody phage display. CARs bearing the CD3ζ and the CD137 costimulatory domains were constructed using these GFRα4-specific scFvs. GFRα4-specific CAR Ts trigger antigen-dependent cytotoxicity and cytokine production in vitro, and they are able to eliminate tumors derived from the MTC TT cell line in an immunodeficient mouse xenograft model of MTC. These data demonstrate the feasibility of targeting GFRα4 by CAR T and support this antigen as a promising target for adoptive T cell immunotherapy and other antibody-based therapies for MTC.

17.
Nat Commun ; 11(1): 6013, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-33243994

RESUMO

SARS-CoV-2 variants with spike (S)-protein D614G mutations now predominate globally. We therefore compare the properties of the mutated S protein (SG614) with the original (SD614). We report here pseudoviruses carrying SG614 enter ACE2-expressing cells more efficiently than those with SD614. This increased entry correlates with less S1-domain shedding and higher S-protein incorporation into the virion. Similar results are obtained with virus-like particles produced with SARS-CoV-2 M, N, E, and S proteins. However, D614G does not alter S-protein binding to ACE2 or neutralization sensitivity of pseudoviruses. Thus, D614G may increase infectivity by assembling more functional S protein into the virion.


Assuntos
COVID-19/virologia , SARS-CoV-2/patogenicidade , Glicoproteína da Espícula de Coronavírus/genética , Vírion/metabolismo , Montagem de Vírus/genética , Internalização do Vírus , Substituição de Aminoácidos , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/epidemiologia , Células HEK293 , Humanos , Mutação , Pandemias , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/metabolismo
18.
bioRxiv ; 2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32637954

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein mediates infection of cells expressing angiotensin-converting enzyme 2 (ACE2). ACE2 is also the viral receptor of SARS-CoV (SARS-CoV-1), a related coronavirus that emerged in 2002-2003. Horseshoe bats (genus Rhinolophus ) are presumed to be the original reservoir of both viruses, and a SARS-like coronavirus, RaTG13, closely related SARS-CoV-2, has been isolated from one horseshoe-bat species. Here we characterize the ability of S-protein receptor-binding domains (RBDs) of SARS-CoV-1, SARS-CoV-2, and RaTG13 to bind a range of ACE2 orthologs. We observed that the SARS-CoV-2 RBD bound human, pangolin, and horseshoe bat ( R. macrotis) ACE2 more efficiently than the SARS-CoV-1 or RaTG13 RBD. Only the RaTG13 RBD bound rodent ACE2 orthologs efficiently. Five mutations drawn from ACE2 orthologs of nine Rhinolophus species enhanced human ACE2 binding to the SARS-CoV-2 RBD and neutralization of SARS-CoV-2 by an immunoadhesin form of human ACE2 (ACE2-Fc). Two of these mutations impaired neutralization of SARS-CoV-1. An ACE2-Fc variant bearing all five mutations neutralized SARS-CoV-2 five-fold more efficiently than human ACE2-Fc. These data narrow the potential SARS-CoV-2 reservoir, suggest that SARS-CoV-1 and -2 originate from distinct bat species, and identify a more potently neutralizing form of ACE2-Fc.

19.
Angew Chem Int Ed Engl ; 59(29): 12178-12185, 2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-32329959

RESUMO

Although macromolecules on cell surfaces are predominantly targeted and drugged with antibodies, they harbor pockets that are only accessible to small molecules and constitutes a rich subset of binding sites with immense potential diagnostic and therapeutic utility. Compared to antibodies, however, small molecules are disadvantaged by a less confined biodistribution, shorter circulatory half-life, and inability to communicate with the immune system. Presented herein is a method that endows small molecules with the ability to recruit and activate chimeric antigen receptor T cells (CAR-Ts). It is based on a CAR-T platform that uses a chemically programmed antibody fragment (cp-Fab) as on/off switch. In proof-of-concept studies, this cp-Fab/CAR-T system targeting folate binding proteins on the cell surface mediated potent and specific eradication of folate-receptor-expressing cancer cells in vitro and in vivo.


Assuntos
Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos , Animais , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Receptor 2 de Folato , Humanos , Camundongos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Superfície Celular , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Biol Chem ; 295(18): 5995-6006, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32193207

RESUMO

Antibodies are widely used as cancer therapeutics, but their current use is limited by the low number of antigens restricted to cancer cells. A receptor tyrosine kinase, receptor tyrosine kinase-like orphan receptor 2 (ROR2), is normally expressed only during embryogenesis and is tightly down-regulated in postnatal healthy tissues. However, it is up-regulated in a diverse set of hematologic and solid malignancies, thus ROR2 represents a candidate antigen for antibody-based cancer therapy. Here we describe the affinity maturation and humanization of a rabbit mAb that binds human and mouse ROR2 but not human ROR1 or other human cell-surface antigens. Co-crystallization of the parental rabbit mAb in complex with the human ROR2 kringle domain (hROR2-Kr) guided affinity maturation by heavy-chain complementarity-determining region 3 (HCDR3)-focused mutagenesis and selection. The affinity-matured rabbit mAb was then humanized by complementarity-determining region (CDR) grafting and framework fine tuning and again co-crystallized with hROR2-Kr. We show that the affinity-matured and humanized mAb retains strong affinity and specificity to ROR2 and, following conversion to a T cell-engaging bispecific antibody, has potent cytotoxicity toward ROR2-expressing cells. We anticipate that this humanized affinity-matured mAb will find application for antibody-based cancer therapy of ROR2-expressing neoplasms.


Assuntos
Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Afinidade de Anticorpos , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/imunologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Especificidade de Anticorpos , Complexo CD3/imunologia , Linhagem Celular Tumoral , Cristalização , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Domínios Proteicos , Coelhos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA