Coronavirus disease 2019 (COVID-19), an infectious disease that primarily attacks the human pulmonary system, is caused by a viral strain called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The outbreak emerged from Wuhan, China, and later spread throughout the world. Until the first week of May 2020, over 3.7 million cases had been reported worldwide and more than 258,000 had died due to the disease. So far, off label use of various drugs has been tried in many clinical settings, however, at present, there is no vaccine or antiviral treatment for human and animal coronaviruses. Therefore, repurposing of the available drugs may be promising to control emerging infections of SARS-COV2; however, new interventions are likely to require months to years to develop. Glycopeptides, which are active against gram-positive bacteria, have demonstrated significant activity against viral infections including SARS-COV and MERS-COV and have a high resemblance of sequence homology with SARS-COV2. Recent in vitro studies have also shown promising activities of aglycon derivative of glycopeptides and teicoplanin against SARS-COV2. Hydrophobic aglycon derivatives and teicoplanin, with minimal toxicity to human cell lines, inhibit entry and replication of SARS-COV2. These drugs block proteolysis of polyprotein a/b with replicase and transcription domains. Teicoplanin use was associated with complete viral clearance in a cohort of patients with severe COVID-19 symptoms. This review attempts to describe the activity, elucidate the possible mechanisms and potential clinical applications of existing glycopeptides against corona viruses, specifically SARS-COV2.
{"title":"Glycopeptides as Potential Interventions for COVID-19.","authors":"Desalegn Getnet Demsie, Abadi Kahsu Gebre, Ebrahim M Yimer, Niguse Meles Alema, Ephrem Mebrahtu Araya, Abere Tilahun Bantie, Mengesha Dessie Allene, Hagazi Gebremedhin, Adane Yehualaw, Chernet Tafere, Haileslassie Tesfay Tadese, Bekalu Amare, Etsay Weldekidan, Desye Gebrie","doi":"10.2147/BTT.S262705","DOIUrl":"https://doi.org/10.2147/BTT.S262705","url":null,"abstract":"<p><p>Coronavirus disease 2019 (COVID-19), an infectious disease that primarily attacks the human pulmonary system, is caused by a viral strain called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The outbreak emerged from Wuhan, China, and later spread throughout the world. Until the first week of May 2020, over 3.7 million cases had been reported worldwide and more than 258,000 had died due to the disease. So far, off label use of various drugs has been tried in many clinical settings, however, at present, there is no vaccine or antiviral treatment for human and animal coronaviruses. Therefore, repurposing of the available drugs may be promising to control emerging infections of SARS-COV2; however, new interventions are likely to require months to years to develop. Glycopeptides, which are active against gram-positive bacteria, have demonstrated significant activity against viral infections including SARS-COV and MERS-COV and have a high resemblance of sequence homology with SARS-COV2. Recent in vitro studies have also shown promising activities of aglycon derivative of glycopeptides and teicoplanin against SARS-COV2. Hydrophobic aglycon derivatives and teicoplanin, with minimal toxicity to human cell lines, inhibit entry and replication of SARS-COV2. These drugs block proteolysis of polyprotein a/b with replicase and transcription domains. Teicoplanin use was associated with complete viral clearance in a cohort of patients with severe COVID-19 symptoms. This review attempts to describe the activity, elucidate the possible mechanisms and potential clinical applications of existing glycopeptides against corona viruses, specifically SARS-COV2.</p>","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"14 ","pages":"107-114"},"PeriodicalIF":4.0,"publicationDate":"2020-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S262705","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10849878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-11-25eCollection Date: 2019-01-01DOI: 10.2147/BTT.S214518
Alok Chandra, Ravi Kanth, Sandeep Thareja
Background: Adalimumab (ADA) is approved for the management of lcerative colitis (UC) not responding to conventional therapy. Use of biologics in resource-constrained settings is very challenging. Currently, real-life data on the safety and efficacy of ADA biosimilar (Exemptia) in steroid-refractory UC patients are limited.
Aim and objectives: To assess the efficacy and safety of ADA biosimilar (Exemptia) to treat steroid-refractory difficult-to-treat UC patients in a resource-constrained Indian setting at 24-weeks follow-up.
Materials and methods: This was a retrospective single-center study to evaluate the efficacy and safety of ADA biosimilar (Exemptia) in steroid-refractory UC patients. All the eligible patients who received induction dose of 160 mg at week 0, 80 mg at week 2 and 40 mg at week 4 and 40 mg every 4 weeks as maintenance regimen from 01 September 2017 to 31 Jan 2019 were retrospectively included in this single-center analysis. Those patients who had shown sub-optimal response at 12 weeks received 40 mg every 2 weeks as maintenance therapy. Outcomes in terms of clinical remission, clinical response and mucosal healing were evaluated in the short term at 12 weeks and 24 weeks.
Results: Twenty-five patients were retrospectively included between the time period of 1 September 2017 to 31 July 2018 with a mean age of 35 years. ADA biosimilar was effective in inducing clinical remission in 16% patients at 12 and 24 weeks, clinical response was seen in 48% at week 12 and 44% at week 24. The mean baseline total Mayo score (TMS) for all patients was 10.16 which decreased to a mean score of 5.72 at 12 weeks and 5.52 at 24 weeks with therapy with the decrease of the score being statistically significant both at 12 and 24 weeks (p<0.05). Two patients (8%) developed pulmonary tuberculosis (TB). ADA biosimilar frequency was accelerated to once in 2 weeks in 14 (56%) patients who did not show an optimal response at 12 weeks. Of these 14 patients, 5 were responders and 9 were non-responders at 12 weeks. At 24 weeks, 6 patients showed clinical response and 7 were non-responders, while one patient had developed TB.
Conclusion: ADA biosimilar (Exemptia) therapy is a safe and cost-effective alternative to original biologics in difficult-to-treat UC patients in resource-constrained Indian setting with comparable efficacy. Maintenance therapy at four weekly intervals can be considered in those patients who have shown an early clinical response at 12 weeks to minimize costs, but more studies are needed to confirm the same.
{"title":"Efficacy And Safety Of Adalimumab Biosimilar (Exemptia) In Moderate-To-Severe Steroid-Refractory Ulcerative Colitis Patients: Real-Life Outcomes In Resource-Constrained Setting At 24-Weeks Follow-Up.","authors":"Alok Chandra, Ravi Kanth, Sandeep Thareja","doi":"10.2147/BTT.S214518","DOIUrl":"10.2147/BTT.S214518","url":null,"abstract":"<p><strong>Background: </strong>Adalimumab (ADA) is approved for the management of lcerative colitis (UC) not responding to conventional therapy. Use of biologics in resource-constrained settings is very challenging. Currently, real-life data on the safety and efficacy of ADA biosimilar (Exemptia) in steroid-refractory UC patients are limited.</p><p><strong>Aim and objectives: </strong>To assess the efficacy and safety of ADA biosimilar (Exemptia) to treat steroid-refractory difficult-to-treat UC patients in a resource-constrained Indian setting at 24-weeks follow-up.</p><p><strong>Materials and methods: </strong>This was a retrospective single-center study to evaluate the efficacy and safety of ADA biosimilar (Exemptia) in steroid-refractory UC patients. All the eligible patients who received induction dose of 160 mg at week 0, 80 mg at week 2 and 40 mg at week 4 and 40 mg every 4 weeks as maintenance regimen from 01 September 2017 to 31 Jan 2019 were retrospectively included in this single-center analysis. Those patients who had shown sub-optimal response at 12 weeks received 40 mg every 2 weeks as maintenance therapy. Outcomes in terms of clinical remission, clinical response and mucosal healing were evaluated in the short term at 12 weeks and 24 weeks.</p><p><strong>Results: </strong>Twenty-five patients were retrospectively included between the time period of 1 September 2017 to 31 July 2018 with a mean age of 35 years. ADA biosimilar was effective in inducing clinical remission in 16% patients at 12 and 24 weeks, clinical response was seen in 48% at week 12 and 44% at week 24. The mean baseline total Mayo score (TMS) for all patients was 10.16 which decreased to a mean score of 5.72 at 12 weeks and 5.52 at 24 weeks with therapy with the decrease of the score being statistically significant both at 12 and 24 weeks (p<0.05). Two patients (8%) developed pulmonary tuberculosis (TB). ADA biosimilar frequency was accelerated to once in 2 weeks in 14 (56%) patients who did not show an optimal response at 12 weeks. Of these 14 patients, 5 were responders and 9 were non-responders at 12 weeks. At 24 weeks, 6 patients showed clinical response and 7 were non-responders, while one patient had developed TB.</p><p><strong>Conclusion: </strong>ADA biosimilar (Exemptia) therapy is a safe and cost-effective alternative to original biologics in difficult-to-treat UC patients in resource-constrained Indian setting with comparable efficacy. Maintenance therapy at four weekly intervals can be considered in those patients who have shown an early clinical response at 12 weeks to minimize costs, but more studies are needed to confirm the same.</p>","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 1","pages":"191-200"},"PeriodicalIF":5.3,"publicationDate":"2019-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6883941/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46186136","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-08-30eCollection Date: 2019-01-01DOI: 10.2147/BTT.S214820
Nur Samsu, Setyawati Soeharto, Muhaimin Rifai, Achmad Rudijanto
Background: Podocyte injury and its subsequent detachment play a critical role in the development and progression of diabetic nephropathy (DN). The objective of this study was to investigate the effect of rosmarinic acid (RA) in preventing podocyte detachment and inhibiting the progression of DN in streptozotocin (STZ)-induced diabetic in rats.
Methods: We used 20 adult male Wistar rats as experimental animals, which were randomly divided into 5 groups (n=4 per group): nondiabetic rat group (negative control) and 4 groups of STZ-induced diabetic rats, namely, 1 group untreated diabetic rats (positive control) and 3 groups treated diabetic rats with RA 75 mg/kg, telmisartan (TMS) 1 mg/kg and combination of RA 75 mg/kg with TMS 1 mg/kg), respectively. After 8 weeks of therapy, urinary levels of podocin, nephrin and albumin and also serum cystatin C levels were examined by ELISA. The expression of p65 nuclear factor-kB by immunohistochemistry whereas expression of podocin and nephrin glomerulus were examined by immunofluorescence.
Results: In the treated diabetic groups, we found that urinary level of podocin and nephrin, albumin urine excretion and serum cystatin C levels were significantly lower than the positive control group. Compared to negative controls, the group of treated diabetic rats did not differ significantly in preventing increased excretion of urinary nephrin and podocin. Meanwhile, treatment with RA monotherapy was significantly better than TMS or a combination of RA with TMS in reducing albumin excretion and preventing decreased kidney function.
Conclusion: In STZ-induced diabetic rats, RA can prevent podocyte detachment. Treatment with RA and TMS either monotherapy or in combination can inhibit the development and progression of DN. However, the combination of both did not show a synergistic effect, even have higher urinary albumin excretion and worse kidney function compared to the RA monotherapy.
{"title":"Rosmarinic acid monotherapy is better than the combination of rosmarinic acid and telmisartan in preventing podocyte detachment and inhibiting the progression of diabetic nephropathy in rats.","authors":"Nur Samsu, Setyawati Soeharto, Muhaimin Rifai, Achmad Rudijanto","doi":"10.2147/BTT.S214820","DOIUrl":"10.2147/BTT.S214820","url":null,"abstract":"<p><strong>Background: </strong>Podocyte injury and its subsequent detachment play a critical role in the development and progression of diabetic nephropathy (DN). The objective of this study was to investigate the effect of rosmarinic acid (RA) in preventing podocyte detachment and inhibiting the progression of DN in streptozotocin (STZ)-induced diabetic in rats.</p><p><strong>Methods: </strong>We used 20 adult male Wistar rats as experimental animals, which were randomly divided into 5 groups (n=4 per group): nondiabetic rat group (negative control) and 4 groups of STZ-induced diabetic rats, namely, 1 group untreated diabetic rats (positive control) and 3 groups treated diabetic rats with RA 75 mg/kg, telmisartan (TMS) 1 mg/kg and combination of RA 75 mg/kg with TMS 1 mg/kg), respectively. After 8 weeks of therapy, urinary levels of podocin, nephrin and albumin and also serum cystatin C levels were examined by ELISA. The expression of p65 nuclear factor-kB by immunohistochemistry whereas expression of podocin and nephrin glomerulus were examined by immunofluorescence.</p><p><strong>Results: </strong>In the treated diabetic groups, we found that urinary level of podocin and nephrin, albumin urine excretion and serum cystatin C levels were significantly lower than the positive control group. Compared to negative controls, the group of treated diabetic rats did not differ significantly in preventing increased excretion of urinary nephrin and podocin. Meanwhile, treatment with RA monotherapy was significantly better than TMS or a combination of RA with TMS in reducing albumin excretion and preventing decreased kidney function.</p><p><strong>Conclusion: </strong>In STZ-induced diabetic rats, RA can prevent podocyte detachment. Treatment with RA and TMS either monotherapy or in combination can inhibit the development and progression of DN. However, the combination of both did not show a synergistic effect, even have higher urinary albumin excretion and worse kidney function compared to the RA monotherapy.</p>","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 ","pages":"179-190"},"PeriodicalIF":5.3,"publicationDate":"2019-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/cb/be/btt-13-179.PMC6722456.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41190498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
R. Melsheimer, A. Geldhof, I. Apaolaza, T. Schaible
Abstract On August 24, 1998, Remicade® (infliximab), the first tumor necrosis factor-α (TNF) inhibitor, received its initial marketing approval from the US Food and Drug Administration for the treatment of Crohn’s disease. Subsequently, Remicade was approved in another five adult and two pediatric indications both in the USA and across the globe. In the 20 years since this first approval, Remicade has made several important contributions to the advancement of science and medicine: 1) clinical trials with Remicade established the proof of concept that targeted therapy can be effective in immune-mediated inflammatory diseases; 2) as the first monoclonal antibody approved for use in a chronic condition, Remicade helped in identifying methods of administering large, foreign proteins repeatedly while limiting the body’s immune response to them; 3) the need to establish Remicade’s safety profile required developing new methods and setting new standards for postmarketing safety studies, specifically in the real-world setting, in terms of approach, size, and duration of follow-up; 4) the study of Remicade has improved our understanding of TNF’s role in the immune system, as well as our understanding of the pathophysiology of a range of diseases characterized by chronic inflammation; and 5) Remicade and other TNF inhibitors have transformed treatment practices in these chronic inflammatory diseases: remission has become a realistic goal of therapy and long-term disability resulting from structural damage can be prevented. This paper reviews how, over the course of its development and 20 years of use in clinical practice, Remicade was able to make these contributions.
{"title":"Remicade® (infliximab): 20 years of contributions to science and medicine","authors":"R. Melsheimer, A. Geldhof, I. Apaolaza, T. Schaible","doi":"10.2147/BTT.S207246","DOIUrl":"https://doi.org/10.2147/BTT.S207246","url":null,"abstract":"Abstract On August 24, 1998, Remicade® (infliximab), the first tumor necrosis factor-α (TNF) inhibitor, received its initial marketing approval from the US Food and Drug Administration for the treatment of Crohn’s disease. Subsequently, Remicade was approved in another five adult and two pediatric indications both in the USA and across the globe. In the 20 years since this first approval, Remicade has made several important contributions to the advancement of science and medicine: 1) clinical trials with Remicade established the proof of concept that targeted therapy can be effective in immune-mediated inflammatory diseases; 2) as the first monoclonal antibody approved for use in a chronic condition, Remicade helped in identifying methods of administering large, foreign proteins repeatedly while limiting the body’s immune response to them; 3) the need to establish Remicade’s safety profile required developing new methods and setting new standards for postmarketing safety studies, specifically in the real-world setting, in terms of approach, size, and duration of follow-up; 4) the study of Remicade has improved our understanding of TNF’s role in the immune system, as well as our understanding of the pathophysiology of a range of diseases characterized by chronic inflammation; and 5) Remicade and other TNF inhibitors have transformed treatment practices in these chronic inflammatory diseases: remission has become a realistic goal of therapy and long-term disability resulting from structural damage can be prevented. This paper reviews how, over the course of its development and 20 years of use in clinical practice, Remicade was able to make these contributions.","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 1","pages":"139 - 178"},"PeriodicalIF":4.0,"publicationDate":"2019-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S207246","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41355401","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}
Pub Date : 2019-07-22eCollection Date: 2019-01-01DOI: 10.2147/BTT.S175034
Dipesh Uprety
Lung cancer is the leading cause of cancer-related mortality worldwide. Non-small-cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases and approximately 70% of patients with NSCLC have locally advanced or metastatic disease at presentation. In NSCLC patients with advanced or metastatic disease, second line treatment with chemotherapy is associated with a poor response rate. In this article, we have reviewed the role of ramucirumab in patients with NSCLC. Ramucirumab is not current standard of care in the first line setting in the treatment of advanced or metastatic NSCLC, based on phase II data which did not show any progression-free survival (PFS) and overall survival (OS) benefit when ramucirumab was compared with non-ramucirumab arm. The REVEL study was a phase III, placebo-controlled trial which included patients with stage IV NSCLC who had progressed during or after platinum-based chemotherapy, with or without bevacizumab. Median OS was 9.1 months vs 10.5 months (HR 0.86, 95% CI 0.75-0.98) in the placebo and ramucirumab group respectively. Seventy-nine percent of patients in ramucirumab arm and 71% of patients in non-ramucirumab arm had grade ≥3 treatment-related adverse events. The addition of ramucirumab to docetaxel can be considered in younger patients with good performance status as a second line treatment option. Additionally, combined blockage of the VEGFR and EGFR pathway has been utilized to overcome resistance to EGFR therapy. The RELAY trial was a phase III, placebo-controlled trial which included patients with sensitizing EGFR mutation positive stage IV NSCLC. Patients were randomized to either ramucirumab plus erlotinib or erlotinib. The trial showed that the combination therapy showed superior PFS benefit.
{"title":"Clinical utility of ramucirumab in non-small-cell lung cancer.","authors":"Dipesh Uprety","doi":"10.2147/BTT.S175034","DOIUrl":"https://doi.org/10.2147/BTT.S175034","url":null,"abstract":"<p><p>Lung cancer is the leading cause of cancer-related mortality worldwide. Non-small-cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases and approximately 70% of patients with NSCLC have locally advanced or metastatic disease at presentation. In NSCLC patients with advanced or metastatic disease, second line treatment with chemotherapy is associated with a poor response rate. In this article, we have reviewed the role of ramucirumab in patients with NSCLC. Ramucirumab is not current standard of care in the first line setting in the treatment of advanced or metastatic NSCLC, based on phase II data which did not show any progression-free survival (PFS) and overall survival (OS) benefit when ramucirumab was compared with non-ramucirumab arm. The REVEL study was a phase III, placebo-controlled trial which included patients with stage IV NSCLC who had progressed during or after platinum-based chemotherapy, with or without bevacizumab. Median OS was 9.1 months vs 10.5 months (HR 0.86, 95% CI 0.75-0.98) in the placebo and ramucirumab group respectively. Seventy-nine percent of patients in ramucirumab arm and 71% of patients in non-ramucirumab arm had grade ≥3 treatment-related adverse events. The addition of ramucirumab to docetaxel can be considered in younger patients with good performance status as a second line treatment option. Additionally, combined blockage of the VEGFR and EGFR pathway has been utilized to overcome resistance to EGFR therapy. The RELAY trial was a phase III, placebo-controlled trial which included patients with sensitizing EGFR mutation positive stage IV NSCLC. Patients were randomized to either ramucirumab plus erlotinib or erlotinib. The trial showed that the combination therapy showed superior PFS benefit.</p>","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 ","pages":"133-137"},"PeriodicalIF":4.0,"publicationDate":"2019-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S175034","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41190497","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Biologics are an important component of the armamentarium of drugs in the treatment of moderate to severe psoriasis. There is increasing evidence that therapeutic drug monitoring (TDM) encompassing the measurement of trough concentrations and anti-drug antibodies (ADA), together with clinical response is emerging as a valuable tool for clinical decision making. It aids in targeted dose adjustments in patients with low drug concentrations, monitoring of adherence and assessment of patients who lose response to biologics or do not respond at all. The high prevalence of psoriasis, its impact on patients’ lives and costs spent on therapy motivate an evidence-based and cost-effective utility of biologics. We performed a literature review on the TDM of TNF alpha antagonists (adalimumab, infliximab, etanercept), IL12/23 antagonists (ustekinumab, guselkumab, tildrakizumab), IL17A inhibitors (secukinumab, ixekizumab) and biosimilars used in the treatment of psoriasis. Although establishing target therapeutic ranges for biologics is ideal, this has only been explored in adalimumab. We also propose a treatment algorithm for the practical application of TDM depending on drug trough concentrations, presence/absence of anti-drug antibodies and clinical response of patients. The practice of TDM is recommended in routine clinical practice where possible.
{"title":"Therapeutic drug monitoring of biologics in psoriasis","authors":"M. M. Liau, H. Oon","doi":"10.2147/BTT.S188286","DOIUrl":"https://doi.org/10.2147/BTT.S188286","url":null,"abstract":"Biologics are an important component of the armamentarium of drugs in the treatment of moderate to severe psoriasis. There is increasing evidence that therapeutic drug monitoring (TDM) encompassing the measurement of trough concentrations and anti-drug antibodies (ADA), together with clinical response is emerging as a valuable tool for clinical decision making. It aids in targeted dose adjustments in patients with low drug concentrations, monitoring of adherence and assessment of patients who lose response to biologics or do not respond at all. The high prevalence of psoriasis, its impact on patients’ lives and costs spent on therapy motivate an evidence-based and cost-effective utility of biologics. We performed a literature review on the TDM of TNF alpha antagonists (adalimumab, infliximab, etanercept), IL12/23 antagonists (ustekinumab, guselkumab, tildrakizumab), IL17A inhibitors (secukinumab, ixekizumab) and biosimilars used in the treatment of psoriasis. Although establishing target therapeutic ranges for biologics is ideal, this has only been explored in adalimumab. We also propose a treatment algorithm for the practical application of TDM depending on drug trough concentrations, presence/absence of anti-drug antibodies and clinical response of patients. The practice of TDM is recommended in routine clinical practice where possible.","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 1","pages":"127 - 132"},"PeriodicalIF":4.0,"publicationDate":"2019-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S188286","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45305554","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}
Objective: The objective of this study was to test our Viral Quinta Columna Strategy (VQCS), a new biological hypothesis predicting that specific multifunctional virally encoded cationic domains may have the capacity to penetrate human cells and interact with PP2A proteins to deregulate important human intracellular pathways, and may display LL37 cathelicidin-like antagonistic effects against multiple pathogens such as bacteria or viruses. Methods: We comparatively analyzed the host defense properties of adenodiaphorins and of some specific cationic sequences encoded by different viruses using two distinct biological models: U87G, a well-characterized cell tumor model; and a group B Streptococcus agalactiae NEM316 ΔdltA, highly sensitive to LL37 cathelicidin. Results: We found that the adenovirus type 2 E4orf4 is a cell-permeable protein containing a new E4orf464–95 protein transduction domain, named large adenodiaphorin or LadD64–95. Interestingly, the host defense LL37 peptide is the unique cathelicidin in humans. In this context, we also demonstrated that similarly to LL37 LadD64–95, several virally encoded cationic sequences including the C-terminus HIV-1 89.6 Vpr77–92, shorter adenodiaphorins AdD67–84/AdD/69–84/AdD69–83, as well as HIV-2 Tat67–90 and JC polyomavirus small t115–134, displayed similar toxicity against Gram-positive S. agalactiae NEM316 ΔdltA strain. Finally, LadD64–95, adenodiaphorin AdD67–84, AdD69–84, and LL37 and LL17–32 cathelicidin peptides also inhibited the survival of human U87G glioblastoma cells. Conclusion: In this study, we demonstrated that specific cationic sequences encoded by four different viruses displayed antibacterial activities against S. agalactiae NEM316 ΔdltA strain. In addition, HIV-1 Vpr71–92 and adenovirus 2 E4orf464–95, two cationic penetrating sequences that bind PP2A, inhibited the survival of U87G glioblastoma cells. These results illustrate the host defense properties of virally encoded sequences and could represent an initial step for future complete validation of the VQCS hypothesis.
{"title":"Antitumor and antibacterial properties of virally encoded cationic sequences","authors":"J. Colle, B. Périchon, Alphonse Garcia","doi":"10.2147/BTT.S201287","DOIUrl":"https://doi.org/10.2147/BTT.S201287","url":null,"abstract":"Objective: The objective of this study was to test our Viral Quinta Columna Strategy (VQCS), a new biological hypothesis predicting that specific multifunctional virally encoded cationic domains may have the capacity to penetrate human cells and interact with PP2A proteins to deregulate important human intracellular pathways, and may display LL37 cathelicidin-like antagonistic effects against multiple pathogens such as bacteria or viruses. Methods: We comparatively analyzed the host defense properties of adenodiaphorins and of some specific cationic sequences encoded by different viruses using two distinct biological models: U87G, a well-characterized cell tumor model; and a group B Streptococcus agalactiae NEM316 ΔdltA, highly sensitive to LL37 cathelicidin. Results: We found that the adenovirus type 2 E4orf4 is a cell-permeable protein containing a new E4orf464–95 protein transduction domain, named large adenodiaphorin or LadD64–95. Interestingly, the host defense LL37 peptide is the unique cathelicidin in humans. In this context, we also demonstrated that similarly to LL37 LadD64–95, several virally encoded cationic sequences including the C-terminus HIV-1 89.6 Vpr77–92, shorter adenodiaphorins AdD67–84/AdD/69–84/AdD69–83, as well as HIV-2 Tat67–90 and JC polyomavirus small t115–134, displayed similar toxicity against Gram-positive S. agalactiae NEM316 ΔdltA strain. Finally, LadD64–95, adenodiaphorin AdD67–84, AdD69–84, and LL37 and LL17–32 cathelicidin peptides also inhibited the survival of human U87G glioblastoma cells. Conclusion: In this study, we demonstrated that specific cationic sequences encoded by four different viruses displayed antibacterial activities against S. agalactiae NEM316 ΔdltA strain. In addition, HIV-1 Vpr71–92 and adenovirus 2 E4orf464–95, two cationic penetrating sequences that bind PP2A, inhibited the survival of U87G glioblastoma cells. These results illustrate the host defense properties of virally encoded sequences and could represent an initial step for future complete validation of the VQCS hypothesis.","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 1","pages":"117 - 126"},"PeriodicalIF":4.0,"publicationDate":"2019-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S201287","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44517857","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}
Butsabong Lerkvaleekul, S. Treepongkaruna, Nichanan Ruangwattanapaisarn, T. Treesit, S. Vilaiyuk
Abstract Systemic polyarteritis nodosa (PAN) is a rare form of necrotizing vasculitis in children. Recurrent episodes of abdominal aneurysm ruptures are uncommon and life-threatening condition in children. Failures of response to immunosuppressive medications and radiological intervention also lead to high mortality. Some reports suggested that tumor necrosis factor (TNF) might have role in the inflammation of this disease. After an English-language literature review, this is the first case report in children of recurrent abdominal-ruptured aneurysms with a failure of conventional therapy but successfully treated with anti-TNF-α monoclonal antibody. We herein describe a 9-year-old girl who presented with chronic abdominal pain, hypertension, and massive lower gastrointestinal bleeding. The disease was refractory to conventional treatment, including administration of a corticosteroid, cyclophosphamide, and intravenous immunoglobulin, and recurrent-ruptured aneurysms developed in the gastrointestinal tract. Arterial embolization during angiography resulted in temporary improvement of the gastrointestinal bleeding. Infliximab, a chimeric anti-tumor necrosis factor-α monoclonal antibody, was initiated and resulted in disease remission with resolution of the gastrointestinal bleeding and abdominal pain. Anti-TNF therapy might be another treatment option for refractory disease to prevent ongoing inflammation that could lead to aneurysmal dilatation or even rupture. However, early recognition of refractory disease and aggressive treatment in the early course of the disease are crucial to reduce morbidity and mortality.
{"title":"Recurrent ruptured abdominal aneurysms in polyarteritis nodosa successfully treated with infliximab","authors":"Butsabong Lerkvaleekul, S. Treepongkaruna, Nichanan Ruangwattanapaisarn, T. Treesit, S. Vilaiyuk","doi":"10.2147/BTT.S204726","DOIUrl":"https://doi.org/10.2147/BTT.S204726","url":null,"abstract":"Abstract Systemic polyarteritis nodosa (PAN) is a rare form of necrotizing vasculitis in children. Recurrent episodes of abdominal aneurysm ruptures are uncommon and life-threatening condition in children. Failures of response to immunosuppressive medications and radiological intervention also lead to high mortality. Some reports suggested that tumor necrosis factor (TNF) might have role in the inflammation of this disease. After an English-language literature review, this is the first case report in children of recurrent abdominal-ruptured aneurysms with a failure of conventional therapy but successfully treated with anti-TNF-α monoclonal antibody. We herein describe a 9-year-old girl who presented with chronic abdominal pain, hypertension, and massive lower gastrointestinal bleeding. The disease was refractory to conventional treatment, including administration of a corticosteroid, cyclophosphamide, and intravenous immunoglobulin, and recurrent-ruptured aneurysms developed in the gastrointestinal tract. Arterial embolization during angiography resulted in temporary improvement of the gastrointestinal bleeding. Infliximab, a chimeric anti-tumor necrosis factor-α monoclonal antibody, was initiated and resulted in disease remission with resolution of the gastrointestinal bleeding and abdominal pain. Anti-TNF therapy might be another treatment option for refractory disease to prevent ongoing inflammation that could lead to aneurysmal dilatation or even rupture. However, early recognition of refractory disease and aggressive treatment in the early course of the disease are crucial to reduce morbidity and mortality.","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 1","pages":"111 - 116"},"PeriodicalIF":4.0,"publicationDate":"2019-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S204726","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41844691","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}
Background: Increasing evidence suggests that TRIP6 has been considered to be aberrantly regulated in several malignancies and involved in tumor growth and metastasis. However, the biological role and prognostic significance of TRIP6 in gastric cancer (GC) still remains unclear. Materials and methods: TRIP6 expression was determined in matched GC tissues and adjacent normal tissues by western blot and real-time PCR. Then, immunohistochemistry was used to detect the expression of TRIP6 in GC patients. Statistical analysis was performed to evaluate the correlation between TRIP6 expression and clinicopathological characteristics and prognosis. Moreover, the effects of TRIP6 on GC cell proliferation and migration were also investigated by using MTT, colony formation and transwell assays. Results: We observed that the expression of TRIP6 was significantly up-regulated in GC tissues and cell ines. Our data indicated that high TRIP6 expression exhibited a significant correlation with poor prognosis for GC patients. Multivariate analysis showed that TRIP6 expression was an independent prognostic factor of the overall survival of GC patients. Furthermore, ectopic expression of TRIP6 promotes cell proliferation and migration in BGC823 cells, whereas knockdown of TRIP6 suppresses cell proliferation and migration in MKN45 cells. Conclusion: These findings demonstrate that TRIP6 exerts an important role in cancer development, which represents a potential prognostic indicator in GC.
{"title":"TRIP6 functions as a potential oncogene and facilitated proliferation and metastasis of gastric cancer","authors":"Lirong Zhu, Xiaodong Xu, Yijie Tang, Xiaochao Zhu","doi":"10.2147/BTT.S191863","DOIUrl":"https://doi.org/10.2147/BTT.S191863","url":null,"abstract":"Background: Increasing evidence suggests that TRIP6 has been considered to be aberrantly regulated in several malignancies and involved in tumor growth and metastasis. However, the biological role and prognostic significance of TRIP6 in gastric cancer (GC) still remains unclear. Materials and methods: TRIP6 expression was determined in matched GC tissues and adjacent normal tissues by western blot and real-time PCR. Then, immunohistochemistry was used to detect the expression of TRIP6 in GC patients. Statistical analysis was performed to evaluate the correlation between TRIP6 expression and clinicopathological characteristics and prognosis. Moreover, the effects of TRIP6 on GC cell proliferation and migration were also investigated by using MTT, colony formation and transwell assays. Results: We observed that the expression of TRIP6 was significantly up-regulated in GC tissues and cell ines. Our data indicated that high TRIP6 expression exhibited a significant correlation with poor prognosis for GC patients. Multivariate analysis showed that TRIP6 expression was an independent prognostic factor of the overall survival of GC patients. Furthermore, ectopic expression of TRIP6 promotes cell proliferation and migration in BGC823 cells, whereas knockdown of TRIP6 suppresses cell proliferation and migration in MKN45 cells. Conclusion: These findings demonstrate that TRIP6 exerts an important role in cancer development, which represents a potential prognostic indicator in GC.","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 1","pages":"101 - 110"},"PeriodicalIF":4.0,"publicationDate":"2019-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S191863","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47849512","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}
We report a case of refractory idiopathic childhood pyoderma gangrenosum in a young boy who had suffered from this disease since 3 years of age. He had unfavorable responses and intermittent relapses under different combinations of cytotoxic and steroid therapies. Although there was not much information available about infliximab use for biologic and childhood pyoderma gangrenosum, eventually we decided to use infliximab in this patient. Infliximab showed a dramatic response and resulted in full recovery during 2 years' follow-up.
{"title":"Infliximab in treatment of idiopathic refractory childhood pyoderma gangrenosum (PG).","authors":"Farhad Salehzadeh, Yusef Mohammadikebar, Afsaneh Enteshary, Omid Ghanbarpour, Mehrdad Mirzarahimi","doi":"10.2147/BTT.S203753","DOIUrl":"https://doi.org/10.2147/BTT.S203753","url":null,"abstract":"<p><p>We report a case of refractory idiopathic childhood pyoderma gangrenosum in a young boy who had suffered from this disease since 3 years of age. He had unfavorable responses and intermittent relapses under different combinations of cytotoxic and steroid therapies. Although there was not much information available about infliximab use for biologic and childhood pyoderma gangrenosum, eventually we decided to use infliximab in this patient. Infliximab showed a dramatic response and resulted in full recovery during 2 years' follow-up.</p>","PeriodicalId":9025,"journal":{"name":"Biologics : Targets & Therapy","volume":"13 ","pages":"97-99"},"PeriodicalIF":4.0,"publicationDate":"2019-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.2147/BTT.S203753","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37367590","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}