{"title":"Correction to: Bringing cell therapy to tumors: considerations for optimal CAR binder design","authors":"","doi":"10.1093/abt/tbae003","DOIUrl":"https://doi.org/10.1093/abt/tbae003","url":null,"abstract":"","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-02-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139963975","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anhye Kim, Min-Seok Oh, Gwan-Ho Lee, Seongeun Song, Mi-sun Byun, Donghoon Choi, Byung-Yong Yu, Howard Lee
Antibody-based therapeutics (ABTs) including monoclonal/polyclonal antibodies and Fc-fusion proteins are increasingly used in disease treatment, driving the global market growth. Understanding the pharmacokinetic (PK) properties of ABTs is crucial for their clinical effectiveness. This study investigated the PK profile and tissue distribution of efineptakin alfa, a long-acting recombinant human interleukin-7 (rhIL-7-hyFc), using enzyme-linked immunosorbent assay (ELISA) and accelerator mass spectrometry (AMS). Total 4 rats were injected intramuscularly with 1 mg/kg of rhIL-7-hyFc containing 14C-rhIL-7-hyFc, prepared via reductive methylation. Serum total radioactivity (TRA) and serum rhIL-7-hyFc concentrations were quantified using AMS and ELISA, respectively. TRA concentrations in organs were determined by AMS. Serum TRA peaked at 10 hours with a terminal half-life of 40 hours. rhIL-7-hyFc exhibited a mean peak concentration at around 17 hours and a rapid elimination with a half-life of 12.3 hours. Peak concentration and area under the curve of TRA were higher than those of rhIL-7-hyFc. Tissue distribution analysis showed elevated TRA concentrations in lymph nodes, kidneys, and spleen, indicating rhIL-7-hyFc’s affinity for these organs. The study also simulated the positions of 14C labeling in rhIL-7-hyFc, identifying specific residues in the fragment of rhIL-7 portion, and provided the explanation of distinct analytes targeted by each method. Combining ELISA and AMS provided advantages by offering sensitivity and specificity for quantification, as well as enabling the identification of analyte forms. The integrated use of ELISA and AMS offer valuable insights for the development and optimization of ABT.
{"title":"Understanding the pharmacokinetic journey of fc-fusion protein, rhIL-7-hyFc, using complementary approach of two analytical methods, accelerator mass spectrometry and ELISA","authors":"Anhye Kim, Min-Seok Oh, Gwan-Ho Lee, Seongeun Song, Mi-sun Byun, Donghoon Choi, Byung-Yong Yu, Howard Lee","doi":"10.1093/abt/tbae004","DOIUrl":"https://doi.org/10.1093/abt/tbae004","url":null,"abstract":"\u0000 \u0000 \u0000 Antibody-based therapeutics (ABTs) including monoclonal/polyclonal antibodies and Fc-fusion proteins are increasingly used in disease treatment, driving the global market growth. Understanding the pharmacokinetic (PK) properties of ABTs is crucial for their clinical effectiveness. This study investigated the PK profile and tissue distribution of efineptakin alfa, a long-acting recombinant human interleukin-7 (rhIL-7-hyFc), using enzyme-linked immunosorbent assay (ELISA) and accelerator mass spectrometry (AMS).\u0000 \u0000 \u0000 \u0000 Total 4 rats were injected intramuscularly with 1 mg/kg of rhIL-7-hyFc containing 14C-rhIL-7-hyFc, prepared via reductive methylation. Serum total radioactivity (TRA) and serum rhIL-7-hyFc concentrations were quantified using AMS and ELISA, respectively. TRA concentrations in organs were determined by AMS.\u0000 \u0000 \u0000 \u0000 Serum TRA peaked at 10 hours with a terminal half-life of 40 hours. rhIL-7-hyFc exhibited a mean peak concentration at around 17 hours and a rapid elimination with a half-life of 12.3 hours. Peak concentration and area under the curve of TRA were higher than those of rhIL-7-hyFc. Tissue distribution analysis showed elevated TRA concentrations in lymph nodes, kidneys, and spleen, indicating rhIL-7-hyFc’s affinity for these organs. The study also simulated the positions of 14C labeling in rhIL-7-hyFc, identifying specific residues in the fragment of rhIL-7 portion, and provided the explanation of distinct analytes targeted by each method.\u0000 \u0000 \u0000 \u0000 Combining ELISA and AMS provided advantages by offering sensitivity and specificity for quantification, as well as enabling the identification of analyte forms. The integrated use of ELISA and AMS offer valuable insights for the development and optimization of ABT.\u0000","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139800525","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anhye Kim, Min-Seok Oh, Gwan-Ho Lee, Seongeun Song, Mi-sun Byun, Donghoon Choi, Byung-Yong Yu, Howard Lee
Antibody-based therapeutics (ABTs) including monoclonal/polyclonal antibodies and Fc-fusion proteins are increasingly used in disease treatment, driving the global market growth. Understanding the pharmacokinetic (PK) properties of ABTs is crucial for their clinical effectiveness. This study investigated the PK profile and tissue distribution of efineptakin alfa, a long-acting recombinant human interleukin-7 (rhIL-7-hyFc), using enzyme-linked immunosorbent assay (ELISA) and accelerator mass spectrometry (AMS). Total 4 rats were injected intramuscularly with 1 mg/kg of rhIL-7-hyFc containing 14C-rhIL-7-hyFc, prepared via reductive methylation. Serum total radioactivity (TRA) and serum rhIL-7-hyFc concentrations were quantified using AMS and ELISA, respectively. TRA concentrations in organs were determined by AMS. Serum TRA peaked at 10 hours with a terminal half-life of 40 hours. rhIL-7-hyFc exhibited a mean peak concentration at around 17 hours and a rapid elimination with a half-life of 12.3 hours. Peak concentration and area under the curve of TRA were higher than those of rhIL-7-hyFc. Tissue distribution analysis showed elevated TRA concentrations in lymph nodes, kidneys, and spleen, indicating rhIL-7-hyFc’s affinity for these organs. The study also simulated the positions of 14C labeling in rhIL-7-hyFc, identifying specific residues in the fragment of rhIL-7 portion, and provided the explanation of distinct analytes targeted by each method. Combining ELISA and AMS provided advantages by offering sensitivity and specificity for quantification, as well as enabling the identification of analyte forms. The integrated use of ELISA and AMS offer valuable insights for the development and optimization of ABT.
{"title":"Understanding the pharmacokinetic journey of fc-fusion protein, rhIL-7-hyFc, using complementary approach of two analytical methods, accelerator mass spectrometry and ELISA","authors":"Anhye Kim, Min-Seok Oh, Gwan-Ho Lee, Seongeun Song, Mi-sun Byun, Donghoon Choi, Byung-Yong Yu, Howard Lee","doi":"10.1093/abt/tbae004","DOIUrl":"https://doi.org/10.1093/abt/tbae004","url":null,"abstract":"\u0000 \u0000 \u0000 Antibody-based therapeutics (ABTs) including monoclonal/polyclonal antibodies and Fc-fusion proteins are increasingly used in disease treatment, driving the global market growth. Understanding the pharmacokinetic (PK) properties of ABTs is crucial for their clinical effectiveness. This study investigated the PK profile and tissue distribution of efineptakin alfa, a long-acting recombinant human interleukin-7 (rhIL-7-hyFc), using enzyme-linked immunosorbent assay (ELISA) and accelerator mass spectrometry (AMS).\u0000 \u0000 \u0000 \u0000 Total 4 rats were injected intramuscularly with 1 mg/kg of rhIL-7-hyFc containing 14C-rhIL-7-hyFc, prepared via reductive methylation. Serum total radioactivity (TRA) and serum rhIL-7-hyFc concentrations were quantified using AMS and ELISA, respectively. TRA concentrations in organs were determined by AMS.\u0000 \u0000 \u0000 \u0000 Serum TRA peaked at 10 hours with a terminal half-life of 40 hours. rhIL-7-hyFc exhibited a mean peak concentration at around 17 hours and a rapid elimination with a half-life of 12.3 hours. Peak concentration and area under the curve of TRA were higher than those of rhIL-7-hyFc. Tissue distribution analysis showed elevated TRA concentrations in lymph nodes, kidneys, and spleen, indicating rhIL-7-hyFc’s affinity for these organs. The study also simulated the positions of 14C labeling in rhIL-7-hyFc, identifying specific residues in the fragment of rhIL-7 portion, and provided the explanation of distinct analytes targeted by each method.\u0000 \u0000 \u0000 \u0000 Combining ELISA and AMS provided advantages by offering sensitivity and specificity for quantification, as well as enabling the identification of analyte forms. The integrated use of ELISA and AMS offer valuable insights for the development and optimization of ABT.\u0000","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139860662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kee Wee Tan, Pengfei Ji, Hang Zhou, Sam Zhang, Weichang Zhou
The COVID-19 pandemic has spurred adoption of revolutionary initiatives by regulatory agencies and pharmaceutical industry worldwide to deliver therapeutic COVID-19 antibodies to patients at unprecedented speed. Among these, timeline of Chemistry, Manufacturing, and Control (CMC), which involves process development and manufacturing activities critical for the assurance of product quality and consistency before first-in-human clinical trials, was greatly reduced from typically 12-15 months (using clonal materials) to approximately 3 months (using non-clonal materials) in multiple cases. In this perspective, we briefly review the acceleration approaches published for therapeutic COVID-19 antibodies and subsequently discuss the applicability of these approaches to achieve investigational new drug (IND) timelines of ≤10 months in over 60 COVID-19 and non-COVID-19 programs performed at WuXi Biologics. We are of the view that, with demonstrated product quality and consistency, innovative approaches used for COVID-19 can be widely applied in all disease areas for greater speed to clinic.
{"title":"Further Accelerating Biologics Development from DNA to IND: The Journey from COVID-19 to Non-COVID-19 Programs","authors":"Kee Wee Tan, Pengfei Ji, Hang Zhou, Sam Zhang, Weichang Zhou","doi":"10.1093/abt/tbae001","DOIUrl":"https://doi.org/10.1093/abt/tbae001","url":null,"abstract":"\u0000 The COVID-19 pandemic has spurred adoption of revolutionary initiatives by regulatory agencies and pharmaceutical industry worldwide to deliver therapeutic COVID-19 antibodies to patients at unprecedented speed. Among these, timeline of Chemistry, Manufacturing, and Control (CMC), which involves process development and manufacturing activities critical for the assurance of product quality and consistency before first-in-human clinical trials, was greatly reduced from typically 12-15 months (using clonal materials) to approximately 3 months (using non-clonal materials) in multiple cases. In this perspective, we briefly review the acceleration approaches published for therapeutic COVID-19 antibodies and subsequently discuss the applicability of these approaches to achieve investigational new drug (IND) timelines of ≤10 months in over 60 COVID-19 and non-COVID-19 programs performed at WuXi Biologics. We are of the view that, with demonstrated product quality and consistency, innovative approaches used for COVID-19 can be widely applied in all disease areas for greater speed to clinic.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139600199","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hui Chen, Sung-Jin Lee, Brian Ouyang, Nicholas Suen, Jay Ye, Cheng-yu Lu, Yang Li
Monoclonal antibodies have been explored in a broad range of applications including receptor agonism. Given the importance of receptor conformation in signaling, the agonistic activity of antibodies that engage these receptors are influenced by many parameters. Tetravalent bispecific antibodies that target the FZD and LRP receptors and subsequently activate WNT signaling have been constructed. Because WNT activation stimulates stem cell proliferation and tissue regeneration, immune effector functions should be eliminated from therapeutic antibodies targeting this pathway. Here, we report an unexpected effect of Fc glycosylation on the agonistic activity of WNT mimetic antibodies. Our findings underscore the importance of antibody format, geometry, and epitope in agonistic antibody design, and highlight the need to establish appropriate early discovery screening strategies to identify hits for further optimization.
{"title":"Effects of fc glycosylation on the activity of WNT mimetic agonistic antibodies","authors":"Hui Chen, Sung-Jin Lee, Brian Ouyang, Nicholas Suen, Jay Ye, Cheng-yu Lu, Yang Li","doi":"10.1093/abt/tbae002","DOIUrl":"https://doi.org/10.1093/abt/tbae002","url":null,"abstract":"\u0000 Monoclonal antibodies have been explored in a broad range of applications including receptor agonism. Given the importance of receptor conformation in signaling, the agonistic activity of antibodies that engage these receptors are influenced by many parameters. Tetravalent bispecific antibodies that target the FZD and LRP receptors and subsequently activate WNT signaling have been constructed. Because WNT activation stimulates stem cell proliferation and tissue regeneration, immune effector functions should be eliminated from therapeutic antibodies targeting this pathway. Here, we report an unexpected effect of Fc glycosylation on the agonistic activity of WNT mimetic antibodies. Our findings underscore the importance of antibody format, geometry, and epitope in agonistic antibody design, and highlight the need to establish appropriate early discovery screening strategies to identify hits for further optimization.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139615189","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Atopic dermatitis (AD) is a chronic inflammatory skin condition characterized by dysregulated immune responses. The key mediators of AD pathogenesis are T helper 2 (TH2) cells and TH2 cytokines. Targeting interleukin 4 (IL4), IL13, or IL31 has become a pivotal focus in both research and clinical treatments for AD. However, the need remains pressing for the development of a more effective and safer therapy, as the current approaches often yield low response rates and adverse effects. In response to this challenge, we have engineered an IgG-scFv (Immunoglobulin G—single-chain Fragment variable) format bispecific antibody designed to concurrently target IL4R and IL31R. Our innovative design involved sequence optimization of VL-VH and the introduction of disulfide bond (VH44-VL100) within the IL31Rα antibody scFv region to stabilize the scFv structure. Our bispecific antibody efficiently inhibited the IL4/IL13/IL31 signaling pathways in vitro and reduced serum Immunoglobulin E (IgE) and IL31 levels in vivo. Consequently, this intervention led to improved inflammation profiles and notable amelioration of AD symptoms. This research highlighted a novel approach to AD therapy by employing bispecific antibody targeting IL4Rα and IL31Rα with potent efficacy.
{"title":"GB12–09, a bispecific antibody targeting IL4Rα and IL31Rα for atopic dermatitis therapy","authors":"Feiyan Deng, Yuxin Qiu, Xiangling Zhang, Nining Guo, Junhong Hu, Wenjie Yang, Wei Shang, Bicheng Liu, Suofu Qin","doi":"10.1093/abt/tbad032","DOIUrl":"https://doi.org/10.1093/abt/tbad032","url":null,"abstract":"\u0000 \u0000 \u0000 Atopic dermatitis (AD) is a chronic inflammatory skin condition characterized by dysregulated immune responses. The key mediators of AD pathogenesis are T helper 2 (TH2) cells and TH2 cytokines. Targeting interleukin 4 (IL4), IL13, or IL31 has become a pivotal focus in both research and clinical treatments for AD. However, the need remains pressing for the development of a more effective and safer therapy, as the current approaches often yield low response rates and adverse effects.\u0000 \u0000 \u0000 \u0000 In response to this challenge, we have engineered an IgG-scFv (Immunoglobulin G—single-chain Fragment variable) format bispecific antibody designed to concurrently target IL4R and IL31R. Our innovative design involved sequence optimization of VL-VH and the introduction of disulfide bond (VH44-VL100) within the IL31Rα antibody scFv region to stabilize the scFv structure.\u0000 \u0000 \u0000 \u0000 Our bispecific antibody efficiently inhibited the IL4/IL13/IL31 signaling pathways in vitro and reduced serum Immunoglobulin E (IgE) and IL31 levels in vivo. Consequently, this intervention led to improved inflammation profiles and notable amelioration of AD symptoms.\u0000 \u0000 \u0000 \u0000 This research highlighted a novel approach to AD therapy by employing bispecific antibody targeting IL4Rα and IL31Rα with potent efficacy.\u0000","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139382968","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xiaozhang Zhang, Ningning Zhou, Chunsheng Yang, Zhaowei Jin, Jeremy Guo
Lyophilized drug products with high protein concentration often perform long reconstitution time, which is inconvenient for clinical use. The objective of this work is to achieve short reconstitution time with multiple and combined strategies. In this paper, we describe the following approaches that lead to reduction of reconstitution time. In terms of lyophilization process, using annealing step during freezing can increase the pore size of lyophilized cake, and decreasing headspace pressure in vial before stoppering can also help solvent penetrate through lyophilized cake. Regarding to formulations, it is an effective strategy of decreasing protein concentration or reducing diluent volume to achieve high protein concentration after reconstitution. Considering container size, high surface-area-to-height ratio of the cakes is also beneficial for reduction of reconstitution time. In addition, reconstitution methods, for example, increasing frequency of swirling and diluent temperature are also helpful to reduce reconstitution time. Among these strategies, reducing diluent volume to achieve high protein concentration and reducing headspace pressure show markedly reduction of reconstitution time. Moreover, we propose combined strategies to mitigate the reconstitution time, at the same time, to achieve same target dose in clinics. The combination of all factors can achieve the overall reduction ratio of 98%, and will not influence the product quality. Therefore, this paper provides insights on the application of multiple strategies to accelerate the reconstitution of lyophilized drug products with high concentration, and facilitates their widespread clinical application.
{"title":"Multiple approaches to reduce reconstitution time of lyophilized drug products with high protein concentration","authors":"Xiaozhang Zhang, Ningning Zhou, Chunsheng Yang, Zhaowei Jin, Jeremy Guo","doi":"10.1093/abt/tbad031","DOIUrl":"https://doi.org/10.1093/abt/tbad031","url":null,"abstract":"Lyophilized drug products with high protein concentration often perform long reconstitution time, which is inconvenient for clinical use. The objective of this work is to achieve short reconstitution time with multiple and combined strategies. In this paper, we describe the following approaches that lead to reduction of reconstitution time. In terms of lyophilization process, using annealing step during freezing can increase the pore size of lyophilized cake, and decreasing headspace pressure in vial before stoppering can also help solvent penetrate through lyophilized cake. Regarding to formulations, it is an effective strategy of decreasing protein concentration or reducing diluent volume to achieve high protein concentration after reconstitution. Considering container size, high surface-area-to-height ratio of the cakes is also beneficial for reduction of reconstitution time. In addition, reconstitution methods, for example, increasing frequency of swirling and diluent temperature are also helpful to reduce reconstitution time. Among these strategies, reducing diluent volume to achieve high protein concentration and reducing headspace pressure show markedly reduction of reconstitution time. Moreover, we propose combined strategies to mitigate the reconstitution time, at the same time, to achieve same target dose in clinics. The combination of all factors can achieve the overall reduction ratio of 98%, and will not influence the product quality. Therefore, this paper provides insights on the application of multiple strategies to accelerate the reconstitution of lyophilized drug products with high concentration, and facilitates their widespread clinical application.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139146021","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
M. Alfaleh, R. Alsulaiman, S. Almahboub, Leena Nezamuldeen, A. Zawawi, Najwa D. Aljehani, Muhammad Yasir, Rwaa H Abdulaal, Rami Alkhaldi, Assala Helal, S. S. Alamri, Jana S. Malki, R. Alhabbab, T. Abujamel, Nabil A Alhakamy, Aisha Alnami, A. Algaissi, Mazen Hassanain, A. Hashem
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and the Middle East respiratory syndrome coronavirus (MERS-CoV) are highly pathogenic human coronaviruses (CoVs). Anti-CoVs mAbs and vaccines may be effective, but the emergence of neutralization escape variants is inevitable. Angiotensin-converting enzyme 2 (ACE2) and dipeptidyl peptidase 4 enzyme (DPP4) are the getaway receptors for SARS-CoV-2 and MERS-CoV, respectively. Thus, we reformatted these receptors and expressed them as recombinant Fc-fusion decoy receptors. Then we tested them in parallel with anti-SARS-CoV (ab1-IgG) and anti-MERS-CoV (M336-IgG) mAbs in ELISA and against several pseudotyped SARS-CoV-2 and MERS-CoV variants using pseudovirus neutralization assay. The generated Fc-based decoy receptors exhibited a strong inhibitory effect against all pseudotyped CoVs. Results showed that although mAbs can be effective antiviral drugs, they might rapidly lose their efficacy against highly mutated viruses, as shown with ab1-IgG against some of the SARS-CoV-2 variants. We suggest that receptor traps can be engineered as Fc-fusion proteins for highly mutating viruses with known entry receptors, for a faster and effective therapeutic response even against virus harboring antibodies escape mutations.
{"title":"ACE2-Fc and DPP4-Fc decoy receptors against SARS-CoV-2 and MERS-CoV variants: A quick therapeutic option for current and future coronaviruses outbreaks","authors":"M. Alfaleh, R. Alsulaiman, S. Almahboub, Leena Nezamuldeen, A. Zawawi, Najwa D. Aljehani, Muhammad Yasir, Rwaa H Abdulaal, Rami Alkhaldi, Assala Helal, S. S. Alamri, Jana S. Malki, R. Alhabbab, T. Abujamel, Nabil A Alhakamy, Aisha Alnami, A. Algaissi, Mazen Hassanain, A. Hashem","doi":"10.1093/abt/tbad030","DOIUrl":"https://doi.org/10.1093/abt/tbad030","url":null,"abstract":"\u0000 \u0000 \u0000 The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and the Middle East respiratory syndrome coronavirus (MERS-CoV) are highly pathogenic human coronaviruses (CoVs). Anti-CoVs mAbs and vaccines may be effective, but the emergence of neutralization escape variants is inevitable.\u0000 \u0000 \u0000 \u0000 Angiotensin-converting enzyme 2 (ACE2) and dipeptidyl peptidase 4 enzyme (DPP4) are the getaway receptors for SARS-CoV-2 and MERS-CoV, respectively. Thus, we reformatted these receptors and expressed them as recombinant Fc-fusion decoy receptors. Then we tested them in parallel with anti-SARS-CoV (ab1-IgG) and anti-MERS-CoV (M336-IgG) mAbs in ELISA and against several pseudotyped SARS-CoV-2 and MERS-CoV variants using pseudovirus neutralization assay.\u0000 \u0000 \u0000 \u0000 The generated Fc-based decoy receptors exhibited a strong inhibitory effect against all pseudotyped CoVs. Results showed that although mAbs can be effective antiviral drugs, they might rapidly lose their efficacy against highly mutated viruses, as shown with ab1-IgG against some of the SARS-CoV-2 variants.\u0000 \u0000 \u0000 \u0000 We suggest that receptor traps can be engineered as Fc-fusion proteins for highly mutating viruses with known entry receptors, for a faster and effective therapeutic response even against virus harboring antibodies escape mutations.\u0000","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138977196","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Daniel Keri, Matt Walker, Isha Singh, Kyle Nishikawa, Fernando Garces
Multispecific antibodies recognize two or more epitopes located on the same or distinct targets. This added capability through protein design allows these man-made molecules to address unmet medical needs that are no longer possible with single targeting such as with monoclonal antibodies or cytokines alone. However, the approach to the development of these multispecific molecules has been met with numerous road bumps, which suggests that a new workflow for multispecific molecules is required. The investigation of the molecular basis that mediates the successful assembly of the building blocks into non-native quaternary structures will lead to the writing of a playbook for multispecifics. This is a must do if we are to design workflows that we can control and in turn predict success. Here, we reflect on the current state-of-the-art of therapeutic biologics and look at the building blocks, in terms of proteins, and tools that can be used to build the foundations of such a next-generation workflow.
{"title":"Next generation of multispecific antibody engineering","authors":"Daniel Keri, Matt Walker, Isha Singh, Kyle Nishikawa, Fernando Garces","doi":"10.1093/abt/tbad027","DOIUrl":"https://doi.org/10.1093/abt/tbad027","url":null,"abstract":"\u0000 Multispecific antibodies recognize two or more epitopes located on the same or distinct targets. This added capability through protein design allows these man-made molecules to address unmet medical needs that are no longer possible with single targeting such as with monoclonal antibodies or cytokines alone. However, the approach to the development of these multispecific molecules has been met with numerous road bumps, which suggests that a new workflow for multispecific molecules is required. The investigation of the molecular basis that mediates the successful assembly of the building blocks into non-native quaternary structures will lead to the writing of a playbook for multispecifics. This is a must do if we are to design workflows that we can control and in turn predict success. Here, we reflect on the current state-of-the-art of therapeutic biologics and look at the building blocks, in terms of proteins, and tools that can be used to build the foundations of such a next-generation workflow.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138588638","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Songna Wang, Pinliang Hu, Jiajun Fan, Jing Zou, Weidong Hong, Xuan Huang, Dan-Ling Pan, Huaning Chen, Yi Zhun Zhu, Liping Ye
The activation of T lymphocytes is a crucial component of the immune response, and the presence of CD80, a membrane antigen, is necessary for T-cell activation. CD80 is usually expressed on antigen-presenting cells (APCs), which can interact with cluster of differentiation 28 (CD28) or programmed cell death ligand 1 (PD-L1) to promote T-cell proliferation, differentiation and function by activating costimulatory signal or blocking inhibitory signal. Simultaneously, CD80 on the APCs also interacts with cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) on the surface of T cells to suppress the response of specific effector T cells, particularly in the context of persistent antigenic stimulation. Due to the pivotal role of CD80 in the immune response, the CD80-Fc fusion protein has emerged as a promising approach for cancer immunotherapy. This review primarily focused on the crucial role of CD80 in the cancer immunotherapy. We also reviewed the current advancements in the research of CD80-Fc fusion proteins. Finally, we deliberated on the challenges encountered by CD80-Fc fusion proteins and proposed the potential strategies that could yield the benefits for patients.
T 淋巴细胞的活化是免疫反应的重要组成部分,而 CD80(一种膜抗原)的存在是 T 细胞活化的必要条件。CD80 通常在抗原递呈细胞(APCs)上表达,可与分化簇 28(CD28)或程序性细胞死亡配体 1(PD-L1)相互作用,通过激活成本刺激信号或阻断抑制信号来促进 T 细胞的增殖、分化和功能。同时,APC 上的 CD80 还与 T 细胞表面的细胞毒性 T 淋巴细胞相关蛋白 4(CTLA-4)相互作用,抑制特定效应 T 细胞的反应,尤其是在持续抗原刺激的情况下。由于 CD80 在免疫反应中的关键作用,CD80-Fc 融合蛋白已成为一种很有前景的癌症免疫疗法。本综述主要关注 CD80 在癌症免疫疗法中的关键作用。我们还回顾了目前 CD80-Fc 融合蛋白研究的进展。最后,我们讨论了 CD80-Fc 融合蛋白所面临的挑战,并提出了可为患者带来益处的潜在策略。
{"title":"CD80-fc fusion protein as a potential cancer immunotherapy strategy","authors":"Songna Wang, Pinliang Hu, Jiajun Fan, Jing Zou, Weidong Hong, Xuan Huang, Dan-Ling Pan, Huaning Chen, Yi Zhun Zhu, Liping Ye","doi":"10.1093/abt/tbad029","DOIUrl":"https://doi.org/10.1093/abt/tbad029","url":null,"abstract":"The activation of T lymphocytes is a crucial component of the immune response, and the presence of CD80, a membrane antigen, is necessary for T-cell activation. CD80 is usually expressed on antigen-presenting cells (APCs), which can interact with cluster of differentiation 28 (CD28) or programmed cell death ligand 1 (PD-L1) to promote T-cell proliferation, differentiation and function by activating costimulatory signal or blocking inhibitory signal. Simultaneously, CD80 on the APCs also interacts with cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) on the surface of T cells to suppress the response of specific effector T cells, particularly in the context of persistent antigenic stimulation. Due to the pivotal role of CD80 in the immune response, the CD80-Fc fusion protein has emerged as a promising approach for cancer immunotherapy. This review primarily focused on the crucial role of CD80 in the cancer immunotherapy. We also reviewed the current advancements in the research of CD80-Fc fusion proteins. Finally, we deliberated on the challenges encountered by CD80-Fc fusion proteins and proposed the potential strategies that could yield the benefits for patients.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139205249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}