1
|
Longmate WM. The epidermal integrin-mediated secretome regulates the skin microenvironment during tumorigenesis and repair. Matrix Biol 2024:S0945-053X(24)00130-6. [PMID: 39491760 DOI: 10.1016/j.matbio.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Integrins are cellular transmembrane receptors that physically connect the cytoskeleton with the extracellular matrix. As such, they are positioned to mediate cellular responses to microenvironmental cues. Importantly, integrins also regulate their own microenvironment through secreted factors, also known as the integrin-mediated secretome. Epidermal integrins, or integrins expressed by keratinocytes of the skin epidermis, regulate the cutaneous microenvironment through the contribution of matrix components, via proteolytic matrix remodeling, or by mediating factors like cytokines and growth factors that can promote support for nearby but distinct cells of the stroma, such as immune cells, endothelial cells, and fibroblasts. This role for integrins is enhanced during both pathological and repair tissue remodeling processes, such as tumor growth and progression and wound healing. This review will discuss examples of how the epithelial integrin-mediated secretome can regulate the tissue microenvironment. Although different epithelial integrins in various contexts will be explored, emphasis will be given to epidermal integrins that regulate the secretome during wound healing and cutaneous tumor progression. Epidermal integrin α3β1 is of particular focus as well, since this integrin has been revealed as a key regulator of the keratinocyte secretome.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
2
|
Bellani S, Molyneaux PL, Maher TM, Spagnolo P. Potential of αvβ6 and αvβ1 integrin inhibition for treatment of idiopathic pulmonary fibrosis. Expert Opin Ther Targets 2024; 28:575-585. [PMID: 38949181 DOI: 10.1080/14728222.2024.2375375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease of unknown cause with a dismal prognosis. Nintedanib and Pirfenidone are approved worldwide for the treatment of IPF, but they only slow the rate of functional decline and disease progression. Therefore, there is an urgent need for more efficacious and better tolerated drugs. AREAS COVERED αvβ6 and αvβ1 are two integrins overexpressed in fibrotic tissue, which play a critical role in the development of lung fibrosis. They act by converting transforming growth factor (TGF)-β, one of the most important profibrotic cytokine, in its active form. Here, we summarize and critically discuss the potential of a dual αvβ6/αvβ1 integrin inhibitor for the treatment of IPF. EXPERT OPINION Bexotegrast, a dual αvβ6/αvβ1 integrin inhibitor, has the potential to slow or even halt disease progression in IPF. Indeed, the strong pre-clinical rationale and promising early phase clinical trial data have raised expectations.
Collapse
Affiliation(s)
- Serena Bellani
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College, London, UK
- Interstitial Lung Disease Unit, Royal Brompton and Harefield Hospitals, London, UK
| | - Toby M Maher
- Hastings Centre for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
3
|
Zhao R, Ding Z, Gupta P, Gozalo L, Bruette R, Johnson VM, Maughn K, Liu Y, Kachroo S. Evaluation of Treatment Patterns and Maintenance Dose Titration Among Patients With Crohn's Disease Initiating Biologics With 3 Years of Follow-Up. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2023; 10:111-120. [PMID: 38025989 PMCID: PMC10664831 DOI: 10.36469/001c.88947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023]
Abstract
Background: There is limited real-world evidence on treatment patterns of patients with Crohn's disease (CD) initiating biologics with an extensive follow-up period. This study describes persistence and dose titration among CD patients with 3 years of follow-up. Methods: This retrospective observational study was conducted using the STATinMED RWD Insights all-payer medical and pharmacy data. Adult patients with at least 1 CD medical claim and at least 1 medical/pharmacy claim for a biologic (adalimumab [ADA], certolizumab pegol (CZP), infliximab [IFX] and its biosimilar products [IFX-BS], ustekinumab [UST], and vedolizumab [VDZ]) between September 2016 and October 2018 were identified. Commercially insured patients with continuous capture for at least 12 months before and at least 36 months after biologics initiation were selected. Confirmed CD patients were included in the final cohort. Baseline patient characteristics and treatment patterns over the 3-year follow-up period were evaluated. Results were summarized using means and SD or counts and percentages. Results: A total of 2309 confirmed patients with CD were identified (847 [36.7%] IFX, 534 [23.1%] ADA, 486 [21.1%] VDZ, 394 [17.1%] UST, 85 [3.7%] CZP, and 72 [3.1%] IFX-BS). CZP and IFX-BS were excluded due to small sample sizes. Approximately half of CD patients were between ages 35 and 54. Patients on UST had a higher Charlson Comorbidity Index score. Common comorbidities (>10%) included anemia, anxiety, depression, and hypertension. Persistence over 3 years' follow-up was highest for UST (61.4%) patients, followed by VDZ (58.0% ), ADA (52.1% , and IFX (48.1%). The discontinuation rate without switch or restart was highest for ADA (37.3%), followed by UST (30.7%), IFX (28.1%), and VDZ (25.3%). Over the 3 years of follow-up, the dose titration rate was highest for IFX (76.5%) and lowest for UST (50.8%). In particular, UST had the lowest dose escalation rate (35.5%) and highest dose-reduction rate (16.5%). Conclusions: Patients with CD on UST had the highest persistence and lowest dose escalation across different biologic users over the 3-year follow-up period, possibly suggesting a better clinical response of UST. Future studies with longer follow-up adjusting for confounders are needed to better understand treatment patterns among biologics users.
Collapse
Affiliation(s)
- Ruizhi Zhao
- Janssen Scientific Affairs, LLC, Horsham, Pennsylvania
| | - Zhijie Ding
- Janssen Scientific Affairs, LLC, Horsham, Pennsylvania
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Abstract
As a natural function, antibodies defend the host from infected cells and pathogens by recognizing their pathogenic determinants. Antibodies (Abs) gained wide acceptance with an enormous impact on human health and have predominantly captured the arena of bio-therapeutics and bio-diagnostics. The scope of Ab-based biologics is vast, and it is likely to solve many unmet clinical needs in future. The majority of attention is now devoted to developing innovative technologies for manufacturing and engineering Abs, better suited to satisfy human needs. The advent of Ab engineering technologies (AET) led to phenomenal developments leading to the generation of Abs-/Ab-derived molecules with desirable functional properties proportional to their expanding requirements. Evolution brought by AET, from the naturally occurring Ab forms to several advanced Ab formats and derivatives, was much needed as it is of great interest to the pharmaceutical industry. Thus, numerous advancements in AET have propelled success in therapeutic Ab development, along with the potential for ever-increasing improvements. Unique characteristics of Abs, such as its diversity, specificity, structural integrity and an array of possible applications, together inspire continuous innovation in the field. Overall, the AET could assist in conquer of several limitations of Abs in terms of their applicability in the field of therapeutics, diagnostics and research; AET has so far led to the production of next-generation Abs, which have revolutionized these arenas. Here in this review, we discuss the various distinguished engineering platforms for Ab development and the progress in modern therapeutics by the so-called "next-generation Abs."
Collapse
Affiliation(s)
- Divya Kandari
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.,Banaras Hindu University, Varanasi, India.,Amity University Rajasthan, Jaipur, India
| |
Collapse
|
5
|
Alorfi NM, Alourfi MM. Biologic Therapy for Refractory Immune Checkpoint Inhibitor Colitis. Biologics 2022; 16:119-127. [PMID: 35957978 PMCID: PMC9362776 DOI: 10.2147/btt.s367675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/08/2022] [Indexed: 11/23/2022]
Abstract
Immune checkpoint inhibitors (ICI) are treatments for several cancer types. Pathogenesis of ICI-induced colitis is not yet clearly explained as it can be disguised as another form such as inflammatory bowel disease or IBD. Recent studies revealed that ICI-induced colitis is a unique form of colitis wherein the synergy of regulatory T cells with the gut microbiome is involved. Diagnosis of colitis can be done via endoscopic lesions and histopathological methods. A patient with colitis can be compared with someone who has IBD. Initial treatment is a corticosteroid. Cooperation between gastroenterologists and oncologists is required to understand further the complete diagnosis and management of different behaviors of ICI-induced colitis. Although immunotherapy provides breakthroughs in treating cancer, adverse effects cannot be prevented and have to be carefully addressed. This study aimed to discuss different biologic therapeutic perspectives in treating refractory immune checkpoint inhibitor-induced colitis. This review provided guidelines, challenges, and suggested protocols for drug immunosuppression.
Collapse
Affiliation(s)
- Nasser M Alorfi
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
- Correspondence: Nasser M Alorfi, Umm Al Qura University, Al Abdeyah, Mecca, 24381, Saudi Arabia, Tel +966500644261, Email
| | - Mansour Marzouq Alourfi
- Internal Medicine Department, King Faisal Medical City for Southern Region, Abha, Saudi Arabia
- Internal Medicine Department, Khamis Mushait General Hospital, Khamis Mushait, Saudi Arabia
| |
Collapse
|
6
|
Vasconcelos D, Chaves B, Albuquerque A, Andrade L, Henriques A, Sartori G, Savino W, Caffarena E, Martins-Da-Silva JH. Development of New Potential Inhibitors of β1 Integrins through In Silico Methods-Screening and Computational Validation. LIFE (BASEL, SWITZERLAND) 2022; 12:life12070932. [PMID: 35888022 PMCID: PMC9325263 DOI: 10.3390/life12070932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022]
Abstract
Integrins are transmembrane receptors that play a critical role in many biological processes which can be therapeutically modulated using integrin blockers, such as peptidomimetic ligands. This work aimed to develop new potential β1 integrin antagonists using modeled receptors based on the aligned crystallographic structures and docked with three lead compounds (BIO1211, BIO5192, and TCS2314), widely known as α4β1 antagonists. Lead-compound complex optimization was performed by keeping intact the carboxylate moiety of the ligand, adding substituents in two other regions of the molecule to increase the affinity with the target. Additionally, pharmacokinetic predictions were performed for the ten best ligands generated, with the lowest docking interaction energy obtained for α4β1 and BIO5192. Results revealed an essential salt bridge between the BIO5192 carboxylate group and the Mg2+ MIDAS ion of the integrin. We then generated more than 200 new BIO5192 derivatives, some with a greater predicted affinity to α4β1. Furthermore, the significance of retaining the pyrrolidine core of the ligand and increasing the therapeutic potential of the new compounds is emphasized. Finally, one novel molecule (1592) was identified as a potential drug candidate, with appropriate pharmacokinetic profiles, similar dynamic behavior at the integrin interaction site compared with BIO5192, and a higher predicted affinity to VLA-4.
Collapse
Affiliation(s)
- Disraeli Vasconcelos
- Laboratório de Biologia Estrutural e Funcional em Biofármacos, Fundação Oswaldo Cruz Ceara, Eusebio 61773-270, Brazil; (D.V.); (B.C.); (A.A.); (L.A.); (A.H.); (G.S.)
| | - Beatriz Chaves
- Laboratório de Biologia Estrutural e Funcional em Biofármacos, Fundação Oswaldo Cruz Ceara, Eusebio 61773-270, Brazil; (D.V.); (B.C.); (A.A.); (L.A.); (A.H.); (G.S.)
| | - Aline Albuquerque
- Laboratório de Biologia Estrutural e Funcional em Biofármacos, Fundação Oswaldo Cruz Ceara, Eusebio 61773-270, Brazil; (D.V.); (B.C.); (A.A.); (L.A.); (A.H.); (G.S.)
| | - Luca Andrade
- Laboratório de Biologia Estrutural e Funcional em Biofármacos, Fundação Oswaldo Cruz Ceara, Eusebio 61773-270, Brazil; (D.V.); (B.C.); (A.A.); (L.A.); (A.H.); (G.S.)
| | - Andrielly Henriques
- Laboratório de Biologia Estrutural e Funcional em Biofármacos, Fundação Oswaldo Cruz Ceara, Eusebio 61773-270, Brazil; (D.V.); (B.C.); (A.A.); (L.A.); (A.H.); (G.S.)
| | - Geraldo Sartori
- Laboratório de Biologia Estrutural e Funcional em Biofármacos, Fundação Oswaldo Cruz Ceara, Eusebio 61773-270, Brazil; (D.V.); (B.C.); (A.A.); (L.A.); (A.H.); (G.S.)
| | - Wilson Savino
- Laboratório de Pesquisa sobre o Timo, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Rede de Pesquisa em Neuroimunomodulação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
| | - Ernesto Caffarena
- Grupo de Biofísica Computacional e Modelagem Molecular, Programa de Computação Científica (PROCC), Fundação Oswaldo Cruz, Rio de Janeiro 21040-222, Brazil;
| | - João Herminio Martins-Da-Silva
- Laboratório de Biologia Estrutural e Funcional em Biofármacos, Fundação Oswaldo Cruz Ceara, Eusebio 61773-270, Brazil; (D.V.); (B.C.); (A.A.); (L.A.); (A.H.); (G.S.)
- Instituto Nacional de Ciência e Tecnologia em Neuroimunomodulação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil
- Correspondence:
| |
Collapse
|
7
|
Bhandari R, Ogeyingbo OD, Kareem R, Gyawali M, Venkatesan N, Ahmed R, Botleroo RA, Elshaikh AO. Efficacy and Safety of Vedolizumab in Management of Moderate to Severe Ulcerative Colitis: A Systematic Review. Cureus 2021; 13:e17729. [PMID: 34659943 PMCID: PMC8491799 DOI: 10.7759/cureus.17729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/04/2021] [Indexed: 01/08/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease and causes inflammation and ulcer of the colon. Vedolizumab is a newer biological agent with an inhibitory effect on α4β7 integrin approved for moderate to severe UC patients. Our study reviewed the clinical response, clinical remission, and mucosal healing of vedolizumab in moderate to severe UC management. Using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we conducted a literature search in PubMed, Google Scholar, and Science Direct, and nine studies were included in the systematic review. At week six, vedolizumab showed a significant clinical response. At week 52, vedolizumab showed significant mucosal healing and clinical remission. The most commonly associated adverse effects are nasopharyngitis, oropharyngeal infection, and gastrointestinal infection. However, additional clinical trials and observational studies with longer follow-ups are required to study the efficacy and safety of the drug.
Collapse
Affiliation(s)
- Renu Bhandari
- Internal Medicine/Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Internal Medicine, Manipal College of Medical Sciences, Kaski, NPL
| | - Opemipo D Ogeyingbo
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Public Health, Walden University, Minneapolis, USA.,Internal Medicine, Saint James School of Medicine, Park Ridge, USA
| | - Roaa Kareem
- Internal Medicine/Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mallika Gyawali
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nanditha Venkatesan
- Internal Medicine, All India Institute of Medical Sciences, Raipur, IND.,Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Rowan Ahmed
- Internal Medicine/Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Rinky A Botleroo
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Abeer O Elshaikh
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
8
|
Blight BJ, Gill AS, Sumsion JS, Pollard CE, Ashby S, Oakley GM, Alt JA, Pulsipher A. Cell Adhesion Molecules are Upregulated and May Drive Inflammation in Chronic Rhinosinusitis with Nasal Polyposis. J Asthma Allergy 2021; 14:585-593. [PMID: 34079296 PMCID: PMC8166329 DOI: 10.2147/jaa.s307197] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/20/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Chronic rhinosinusitis with nasal polyps (CRSwNP) is predominantly characterized by eosinophil- and T helper 2 cell (Th2)-biased inflammation. Integrins and intercellular adhesion molecules (ICAMs) are superfamilies of cell adhesion molecules (CAMs) that facilitate the recruitment and trafficking of immune cells and have been implicated in coordinating eosinophil and Th2 cell adhesion and signaling in asthma. The roles of CAMs in CRSwNP, however, remain poorly understood. The purpose of this study was to characterize the systemic and local expression of CAMs and identify which CAMs are potentially involved in CRSwNP pathology. Materials and Methods A prospective observational study was conducted using peripheral blood and anterior ethmoid tissues of patients with CRSwNP (n=32) and controls (n=15). Multiplex gene analysis and Pearson correlations were performed to identify associations between systemically and locally expressed CAMs. Based on the gene expression results, immunohistochemical evaluation and quantification of cells expressing integrins ITGAM, ITGAX, and ITGB2, as well as ICAM-3 in sinonasal tissues were conducted to compare local protein expression patterns. Results Integrin and ICAM genes were significantly elevated in the blood (p<0.001 to p<0.05) and sinuses (p<0.0003 to p<0.05) of patients with CRSwNP compared to controls. Strong positive correlations of genes expressed in the blood (p<0.01 to p<0.05) and sinuses (p<0.01) were observed between ITGAM, ITGAX, ITGB2, and ICAM3. ITGAM-, ITGB2-, ICAM-3-, and ICAM-3/ITGB2-positive cell counts were significantly increased in CRSwNP compared to controls (p<0.0001 to p<0.04), and a positive correlation between ICAM-3/ITGB2- and ITGAM-positive cell counts was observed (p<0.02). Conclusion The systemic and local expression of ITGAM, ITGB2, and ICAM-3 is significantly upregulated in CRSwNP, suggesting that integrin complex ITGAM/ITGB2 and ICAM-3 serve a potential role in inflammation-mediated signaling in CRSwNP.
Collapse
Affiliation(s)
- Brennan J Blight
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Amarbir S Gill
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jorgen S Sumsion
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Chelsea E Pollard
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Shaelene Ashby
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Gretchen M Oakley
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Jeremiah A Alt
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Abigail Pulsipher
- Division of Otolaryngology-Head and Neck Surgery, Rhinology-Sinus and Skull Base Surgery Program, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
9
|
Truffi M, Sevieri M, Morelli L, Monieri M, Mazzucchelli S, Sorrentino L, Allevi R, Bonizzi A, Zerbi P, Marchini B, Longhi E, Sampietro GM, Colombo F, Prosperi D, Colombo M, Corsi F. Anti-MAdCAM-1-Conjugated Nanocarriers Delivering Quantum Dots Enable Specific Imaging of Inflammatory Bowel Disease. Int J Nanomedicine 2020; 15:8537-8552. [PMID: 33173291 PMCID: PMC7646444 DOI: 10.2147/ijn.s264513] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Assessment of inflammatory bowel disease (IBD) currently relies on aspecific clinical signs of bowel inflammation. Specific imaging of the diseased bowel regions is still lacking. Here, we investigate mucosal addressin cell adhesion molecule 1 (MAdCAM-1) as a reliable and specific endothelial target for engineered nanoparticles delivering imaging agents to obtain an exact mapping of diseased bowel foci. Materials and Methods We generated a nanodevice composed of PLGA-PEG coupled with anti-MAdCAM-1 antibody half-chains and loaded with quantum dots (P@QD-MdC NPs). Bowel localization and systemic biodistribution of the nanoconjugate were analyzed upon injection in a murine model of chronic IBD obtained through repeated administration of dextran sulfate sodium salt. Specificity for diseased bowel regions was also assessed ex vivo in human specimens from patients with IBD. Potential for development as contrast agent in magnetic resonance imaging was assessed by preliminary study on animal model. Results Synthesized nanoparticles revealed good stability and monodispersity. Molecular targeting properties were analyzed in vitro in a cell culture model. Upon intravenous injection, P@QD-MdC NPs were localized in the bowel of colitic mice, with enhanced accumulation at 24 h post-injection compared to untargeted nanoparticles (p<0.05). Nanoparticles injection did not induce histologic lesions in non-target organs. Ex vivo exposure of human bowel specimens to P@QD-MdC NPs revealed specific recognition of the diseased regions vs uninvolved tracts (p<0.0001). After loading with appropriate contrast agent, the nanoparticles enabled localized contrast enhancement of bowel mucosa in the rectum of treated mice. Conclusion P@QD-MdC NPs efficiently detected bowel inflammation foci, accurately following the expression pattern of MAdCAM-1. Fine-tuning of this nanoconjugate with appropriate imaging agents offers a promising non-invasive tool for specific IBD diagnosis.
Collapse
Affiliation(s)
- Marta Truffi
- Nanomedicine and Molecular Imaging Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, Pavia 27100, Italy
| | - Marta Sevieri
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Lucia Morelli
- NanoBioLab, Department of Biotechnology and Biosciences, Università degli studi di Milano-Bicocca, Milano, 20126, Italy
| | - Matteo Monieri
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Serena Mazzucchelli
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Luca Sorrentino
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Raffaele Allevi
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Arianna Bonizzi
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Pietro Zerbi
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Beatrice Marchini
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Erika Longhi
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy
| | - Gianluca Matteo Sampietro
- IBD Surgery Unit, ASST Fatebenefratelli Sacco - Ospedale "Luigi Sacco" Polo Universitario, Milano, 20157, Italy
| | - Francesco Colombo
- IBD Surgery Unit, ASST Fatebenefratelli Sacco - Ospedale "Luigi Sacco" Polo Universitario, Milano, 20157, Italy
| | - Davide Prosperi
- NanoBioLab, Department of Biotechnology and Biosciences, Università degli studi di Milano-Bicocca, Milano, 20126, Italy
| | - Miriam Colombo
- NanoBioLab, Department of Biotechnology and Biosciences, Università degli studi di Milano-Bicocca, Milano, 20126, Italy
| | - Fabio Corsi
- Department of Biomedical and Clinical Sciences "Luigi Sacco, Università degli studi di Milano, Milano, 20157, Italy.,Surgery Department, Breast Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, 27100, Italy
| |
Collapse
|
10
|
Mishra R, Dhawan P, Srivastava AS, Singh AB. Inflammatory bowel disease: Therapeutic limitations and prospective of the stem cell therapy. World J Stem Cells 2020; 12:1050-1066. [PMID: 33178391 PMCID: PMC7596447 DOI: 10.4252/wjsc.v12.i10.1050] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/02/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD), consisting primarily of ulcerative colitis and Crohn’s disease, is a group of debilitating auto-immune disorders, which also increases the risk of colitis-associated cancer. However, due to the chronic nature of the disease and inconsistent treatment outcomes of current anti-IBD drugs (e.g., approximately 30% non-responders to anti-TNFα agents), and related serious side effects, about half of all IBD patients (in millions) turn to alternative treatment options. In this regard, mucosal healing is gaining acceptance as a measure of disease activity in IBD patients as recent studies have correlated the success of mucosal healing with improved prognosis. However, despite the increasing clinical realization of the significance of the concept of mucosal healing, its regulation and means of therapeutic targeting remain largely unclear. Here, stem-cell therapy, which uses hematopoietic stem cells or mesenchymal stem cells, remains a promising option. Stem cells are the pluripotent cells with ability to differentiate into the epithelial and/or immune-modulatory cells. The over-reaching concept is that the stem cells can migrate to the damaged areas of the intestine to provide curative help in the mucosal healing process. Moreover, by differentiating into the mature intestinal epithelial cells, the stem cells also help in restoring the barrier integrity of the intestinal lining and hence prevent the immunomodulatory induction, the root cause of the IBD. In this article, we elaborate upon the current status of the clinical management of IBD and potential role of the stem cell therapy in improving IBD therapy and patient’s quality of life.
Collapse
Affiliation(s)
- Rangnath Mishra
- Global Institute of Stem Cell Therapy and Research, San Diego, CA 92122, United States
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68118, United States
- Fred and Pamela Buffett Cancer Center, Omaha, NE 68118, United States
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68118, United States
| | - Anand S Srivastava
- Global Institute of Stem Cell Therapy and Research, San Diego, CA 92122, United States
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68118, United States
- Fred and Pamela Buffett Cancer Center, Omaha, NE 68118, United States
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68118, United States
| |
Collapse
|
11
|
Yao J, Gao R, Luo M, Li D, Guo L, Yu Z, Xiong F, Wei C, Wu B, Xu Z, Zhang D, Wang J, Wang L. Close homolog of L1-deficient ameliorates inflammatory bowel disease by regulating the balance of Th17/Treg. Gene 2020; 757:144931. [PMID: 32640308 DOI: 10.1016/j.gene.2020.144931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The aim of this study is to investigate the role of close homolog of L1 (CHL1) on inflammatory bowel disease (IBD), and the correlation with the balance of Th17/Treg. METHODS Dextran sodium sulfate (DSS)-induced IBD mice model was established. CHL1 knockout (KO) mice and CHL1 wild-type (WT) mice were subjected to DSS. CHL1 expression was detected using qRT-PCR. Weight was recorded daily, and disease activity index (DAI) score was assessed. The colon length and histological changes were measured. The number of neutrophils, macrophages and T cells was observed by immunohistochemistry. The expression of inflammatory cytokines and the proportion of Th17/Treg cells were detected by qRT-PCR and flow cytometry. The expression of RORγt, STAT3 and Foxp3 was detected by using immunohistochemistry and Western blot. RESULTS CHL1 expression was upregulated in DSS-induced IBD mice. DSS-CHLl-KO mice exhibited less weight loss than the DSS-CHLl-WT mice. The DAI score and histological score were decreased in DSS-CHLl-KO mice compared with DSS-CHLl-WT mice, while colon length was increased. Number of neutrophils, macrophages and T cells, and expression of TNF-α, IL-6, IL-17A, IL-21 and IL-23 were decreased in DSS-CHLl-KO mice, while IL-10 expression was increased. Moreover, CHL1-deficient inhibited Th17 cells differentiation and promoted Treg cells differentiation in IBD mice. CHL1-deficient also inhibited the expression of RORγt and STAT3, and promoted the expression of Foxp3 in IBD mice. CONCLUSION CHL1-deficient reduces the inflammatory response by regulating the balance of Th17/Treg in mice with IBD. CHL1 is expected to be a new target for the treatment of IBD.
Collapse
Affiliation(s)
- Jun Yao
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Ruoyu Gao
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Minghan Luo
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Defeng Li
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Liliangzi Guo
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Zichao Yu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Feng Xiong
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Cheng Wei
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Benhua Wu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Zhenglei Xu
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| | - Dingguo Zhang
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China.
| | - Jianyao Wang
- Department of General Surgery, Shenzhen Children's Hospital, No. 7019, Yitian Road, Shenzhen City, Guangdong Province 518026, China
| | - Lisheng Wang
- Department of Gastroenterology, Jinan University of Second Clinical Medical Sciences, Shenzhen Municipal People's Hospital, No. 1017, East Gate Road, Shenzhen City, Guangdong Province 518020, China
| |
Collapse
|
12
|
Yang S, Arrode-Bruses G, Frank I, Grasperge B, Blanchard J, Gettie A, Martinelli E, Ho EA. Anti-α 4β 7 monoclonal antibody-conjugated nanoparticles block integrin α 4β 7 on intravaginal T cells in rhesus macaques. SCIENCE ADVANCES 2020; 6:6/34/eabb9853. [PMID: 32937372 PMCID: PMC7442472 DOI: 10.1126/sciadv.abb9853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/10/2020] [Indexed: 06/11/2023]
Abstract
Intravenous administration of anti-α4β7 monoclonal antibody in macaques decreases simian immunodeficiency virus (SIV) vaginal infection and reduces gut SIV loads. Because of potential side effects of systemic administration, a prophylactic strategy based on mucosal administration of anti-α4β7 antibody may be safer and more effective. With this in mind, we developed a novel intravaginal formulation consisting of anti-α4β7 monoclonal antibody-conjugated nanoparticles (NPs) loaded in a 1% hydroxyethylcellulose (HEC) gel (NP-α4β7 gel). When intravaginally administered as a single dose in a rhesus macaque model, the formulation preferentially bound to CD4+ or CD3+ T cells expressing high levels of α4β7, and occupied ~40% of α4β7 expressed by these subsets and ~25% of all cells expressing α4β7 Blocking of the α4β7 was restricted to the vaginal tract without any changes detected systemically.
Collapse
Affiliation(s)
- Sidi Yang
- School of Pharmacy, University of Waterloo, 10 Victoria St. S A, Kitchener, Ontario N2G 1C5, Canada
| | - Geraldine Arrode-Bruses
- Center for Biomedical Research, Population Council, One Dag Hammarskjold Plaza, New York, NY 10017, USA
| | - Ines Frank
- Center for Biomedical Research, Population Council, One Dag Hammarskjold Plaza, New York, NY 10017, USA
| | - Brooke Grasperge
- Tulane National Primate Research Center, Tulane University, 6823 St. Charles Ave., New Orleans, LA 70118, USA
| | - James Blanchard
- Tulane National Primate Research Center, Tulane University, 6823 St. Charles Ave., New Orleans, LA 70118, USA
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, Rockefeller University, 455 1st Avenue #7, New York, NY 10016, USA
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, One Dag Hammarskjold Plaza, New York, NY 10017, USA.
| | - Emmanuel A Ho
- School of Pharmacy, University of Waterloo, 10 Victoria St. S A, Kitchener, Ontario N2G 1C5, Canada.
| |
Collapse
|
13
|
Martins F, Sykiotis GP, Maillard M, Fraga M, Ribi C, Kuntzer T, Michielin O, Peters S, Coukos G, Spertini F, Thompson JA, Obeid M. New therapeutic perspectives to manage refractory immune checkpoint-related toxicities. Lancet Oncol 2020; 20:e54-e64. [PMID: 30614479 DOI: 10.1016/s1470-2045(18)30828-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/09/2018] [Accepted: 10/30/2018] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors are reshaping the prognosis of many cancer and are progressively becoming the standard of care in the treatment of many tumour types. Immunotherapy is bringing new hope to patients, but also a whole new spectrum of toxicities for healthcare practitioners to manage. Oncologists and specialists involved in the pluridisciplinary management of patients with cancer are increasingly confronted with the therapeutic challenge of treating patients with severe and refractory immune-related adverse events. In this Personal View, we summarise the therapeutic strategies that have been used to manage such toxicities resulting from immune checkpoint inhibitor treatment. On the basis of current knowledge about their pathogenesis, we discuss the use of new biological and non-biological immunosuppressive drugs to treat severe and steroid refractory immune-related adverse events. Depending on the immune infiltrate type that is predominant, we propose a treatment algorithm for personalised management that goes beyond typical corticosteroid use. We propose a so-called shut-off strategy that aims at inhibiting key inflammatory components involved in the pathophysiological processes of immune-related adverse events, and limits potential adverse effects of drug immunosuppression on tumour response. This approach develops on current guidelines and challenges the step-by-step increase approach to drug immunosuppression.
Collapse
Affiliation(s)
- Filipe Martins
- Département d'Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Gerasimos P Sykiotis
- Service d'Endocrinologie, Diabétologie, et Métabolisme, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Michel Maillard
- Service de Gastro-entérologie et Hépatologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Crohn's and Colitis Center, Lausanne, Switzerland
| | - Montserrat Fraga
- Service de Gastro-entérologie et Hépatologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Camillo Ribi
- Service Immunologie et Allergie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Thierry Kuntzer
- Service de Neurologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Olivier Michielin
- Département d'Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Solange Peters
- Département d'Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Georges Coukos
- Département d'Oncologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Epalinges, Switzerland
| | - Francois Spertini
- Service Immunologie et Allergie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - John A Thompson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA; National Cancer Institute, Bethesda, MA, USA
| | - Michel Obeid
- Service Immunologie et Allergie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Centre d'Immunothérapie et de Vaccinologie, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Faculté Médecine Paris Descartes, Université Paris Descartes, Paris, France.
| |
Collapse
|
14
|
Babbar A, Hitch TCA, Pabst O, Clavel T, Hübel J, Eswaran S, Wagner N, Schippers A. The Compromised Mucosal Immune System of β7 Integrin-Deficient Mice Has Only Minor Effects on the Fecal Microbiota in Homeostasis. Front Microbiol 2019; 10:2284. [PMID: 31636620 PMCID: PMC6787405 DOI: 10.3389/fmicb.2019.02284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract is an ideal habitat for diverse bacterial species that reside in a homeostatic balance with local tissue and significantly contribute to host health. Negative shifts in gut microbiota profiles, also known as dysbiosis, may be implicated in the development of chronic disorders such as inflammatory bowel diseases (IBD). Adhesion molecule-dependent recruitment of immune cells to the gut is an important step in IBD pathogenesis. The adhesion molecule β7 integrin contributes to the development of the gut-associated lymphoid tissue (GALT), intestinal immune cell homing, and immune responses and is known to promote intestinal inflammation. Although many studies underlined the role of the gut microbiota in shaping the mucosal immune system, studies on the influence of the host immune system on the microbiota are rare, especially in homeostasis. We addressed this question via comparative 16S rRNA gene amplicon analysis of fecal microbial communities from wild-type and β7 integrin-deficient mice, the latter being characterized by a compromised GALT. Besides subtle changes in relative abundances of Muribaculaceae spp. and unknown members of the families Ruminococcaceae and Lachnospiraceae, there was altogether no major difference in microbiota profiles in β7 integrin-deficient mice vs. wild-type littermates. This indicates that, in conditions of homeostasis, there is only a minor influence of the host immune system on the fecal microbiota in our mouse model, stressing the potential importance of pathological factors for dysbiosis development.
Collapse
Affiliation(s)
- Anshu Babbar
- Department of Pediatrics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Jessica Hübel
- Department of Pediatrics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Sreepradha Eswaran
- Department of Pediatrics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Norbert Wagner
- Department of Pediatrics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Angela Schippers
- Department of Pediatrics, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
15
|
|
16
|
Wen T, Aronow BJ, Rochman Y, Rochman M, Kc K, Dexheimer PJ, Putnam P, Mukkada V, Foote H, Rehn K, Darko S, Douek D, Rothenberg ME. Single-cell RNA sequencing identifies inflammatory tissue T cells in eosinophilic esophagitis. J Clin Invest 2019; 129:2014-2028. [PMID: 30958799 DOI: 10.1172/jci125917] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/21/2019] [Indexed: 12/26/2022] Open
Abstract
T cell heterogeneity is highly relevant to allergic disorders. We resolved the heterogeneity of human tissue CD3+ T cells during allergic inflammation, focusing on a tissue-specific allergic disease, eosinophilic esophagitis (EoE). We investigated 1088 single T cells derived from patients with a spectrum of disease activity. Eight disparate tissue T cell subtypes (designated T1-T8) were identified, with T7 and T8 enriched in the diseased tissue. The phenotypes of T7 and T8 resemble putative Treg (FOXP3+) and effector Th2-like (GATA3+) cells, respectively. Prodigious levels of IL-5 and IL-13 were confined to HPGDS+ CRTH2+IL-17RB+FFAR3+CD4+ T8 effector Th2 cells. EoE severity closely paralleled a lipid/fatty acid-induced activation node highlighted by the expression of the short-chain fatty acid receptor FFAR3. Ligands for FFAR3 induced Th2 cytokine production from human and murine T cells, including in an in vivo allergy model. Therefore, we elucidated the defining characteristics of tissue-residing CD3+ T cells in EoE, a specific enrichment of CD4+ Treg and effector Th2 cells, confinement of type 2 cytokine production to the CD4+ effector population, a highly likely role for FFAR3 in amplifying local Th2 responses in EoE, and a resource to further dissect tissue lymphocytes and allergic responses.
Collapse
Affiliation(s)
- Ting Wen
- Division of Allergy and Immunology
| | | | | | | | - Kiran Kc
- Division of Allergy and Immunology
| | | | - Philip Putnam
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Vincent Mukkada
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | | - Sam Darko
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
17
|
Yoo SM, Chung SH. Targets of monoclonal antibodies for immunological diseases. Arch Pharm Res 2018; 42:293-304. [PMID: 30426387 DOI: 10.1007/s12272-018-1087-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/31/2018] [Indexed: 01/28/2023]
Abstract
Immunological disorders such as allergy, autoimmune diseases, auto-inflammatory syndromes and immunological deficiency syndromes are difficult to treat with chemical drugs. Recently, many monoclonal antibodies targeting various molecules including interleukin, tumor necrosis factor-α, cluster of differentiation, integrins, complement C5 and B lymphocyte stimulator are clinically available and give a hope to patients suffering from these intractable diseases. Here, we selected twenty-seven monoclonal antibodies approved by US FDA since 1997 and they are classified according to their target molecules. Although these biomedicines possessed some restrictions such as high cost and susceptible to infectious disease, these drawbacks can be overcome through cost-cutting innovations including biosimilars and careful monitoring. New targets are emerging rapidly and more effective biomedicines with acceptable side effects are in the pipeline for next decade.
Collapse
Affiliation(s)
- Seon Min Yoo
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Sung Hyun Chung
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea.
| |
Collapse
|
18
|
Allamneni C, Venkata K, Yun H, Xie F, DeLoach L, Malik TA. Comparative Effectiveness of Vedolizumab vs. Infliximab Induction Therapy in Ulcerative Colitis: Experience of a Real-World Cohort at a Tertiary Inflammatory Bowel Disease Center. Gastroenterology Res 2018; 11:41-45. [PMID: 29511405 PMCID: PMC5827901 DOI: 10.14740/gr934w] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 11/29/2017] [Indexed: 01/02/2023] Open
Abstract
Background Vedolizumab (VDZ), an adhesion molecule inhibitor and infliximab (IFX), a tumor necrosis factor (TNF) blocker, are both approved as first-line induction agents in moderately to severely active ulcerative colitis (UC). However, there are no head-to-head studies comparing the relative effectiveness of the two agents. Here we provide a real-world comparison of these two agents. Methods We conducted an ambidirectional cohort study of adult UC patients seen at our tertiary inflammatory bowel disease (IBD) center from 2012 to 2017. Each patient had moderately to severely active UC via partial Mayo score and was induced with IFX or VDZ. They were followed until assessment of clinical response. Poisson regression was used to calculate clinical response rates and rate ratios. Results Of 59 patients who met inclusion criteria, 27 and 32 patients were induced with IFX and VDZ, respectively. Totally, 18/27 (66.7%) patients induced with IFX vs. 24/32 (78.1%) patients induced with VDZ were clinical responders. Response rates per 100 person-weeks (PW) were similar for VDZ (5.21) and IFX (5.38). The effectiveness in terms of induction of clinical response (incidence rate ratio, IRR) was not statistically significant for VDZ vs. IFX (IRR 0.97, 95% confidence interval (CI) 0.53 - 1.77). Among TNF blocker naive patients, IRR was also not statistically significant between VDZ (6.74/100 PW) and IFX (6.48/100 PW) (IRR 1.04, 95% CI 0.47 - 2.29). Among TNF blocker experienced patients, there was a higher response rate for VDZ (4.52) vs. IFX (2.29) per 100 PW, but the IRR did not reveal statistical significance (IRR 1.97, 95% CI 0.45 - 8.63) due to small sample size of TNF blocker experienced patients who received IFX. Five patients developed severe infection or adverse reaction during IFX induction requiring exclusion, whereas no VDZ patients were excluded for this reason. Conclusions Our study revealed a higher proportion of patients who responded to VDZ vs. IFX; however when accounting for period between induction and assessment of clinical response, rates of clinical response were similar. A key difference between the two groups was the higher response rate in the VDZ group among TNF blocker experienced patients; however, a larger cohort is needed to further elaborate on this difference. VDZ held its own against IFX and this study strengthens its standing as a first-line agent among TNF blocker naive as well as TNF blocker experienced UC patients.
Collapse
Affiliation(s)
- Chaitanya Allamneni
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Krishna Venkata
- Department of Medicine, University of Alabama at Birmingham, Montgomery, AL 36116, USA
| | - Huifeng Yun
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Fenglong Xie
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lindsey DeLoach
- Department of Pharmacy, University of Alabama at Birmingham, Birmingham, AL 35249, USA
| | - Talha A Malik
- Department of Medicine-Gastroenterology, University of Alabama at Birmingham, Birmingham, AL 35249, USA
| |
Collapse
|
19
|
Hassan-Zahraee M, Banerjee A, Cheng JB, Zhang W, Ahmad A, Page K, von Schack D, Zhang B, Martin SW, Nayak S, Reddy P, Xi L, Neubert H, Fernandez Ocana M, Gorelick K, Clare R, Vincent M, Cataldi F, Hung K. Anti-MAdCAM Antibody Increases ß7+ T Cells and CCR9 Gene Expression in the Peripheral Blood of Patients With Crohn's Disease. J Crohns Colitis 2018; 12:77-86. [PMID: 28961803 PMCID: PMC5881777 DOI: 10.1093/ecco-jcc/jjx121] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/01/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To define pharmacodynamic biomarkers in the peripheral blood of patients with Crohn's disease [CD] after treatment with PF-00547659, an anti-human mucosal addressin cell adhesion molecule-1 [MAdCAM-1] monoclonal antibody. METHODS In this Phase 2, randomised, double-blind, controlled study [OPERA], blood samples were analysed from patients with moderate to severe active CD who received placebo or 22.5 mg, 75 mg, or 225 mg of PF-00547659 subcutaneously at baseline and at Weeks 4 and 8, with follow-up at Week 12. Soluble MAdCAM [sMAdCAM] was measured by mass spectrometry, β7-expressing T cells by flow cytometry, and gene transcriptome by RNA sequencing. RESULTS A slight increase in sMAdCAM was measured in the placebo group from baseline to Week 12 [6%], compared with significant decreases in all PF-00547659 groups [-87% to -98%]. A slight increase from baseline to Week 12 was observed in frequency and molecules of equivalent soluble fluorochrome for β7+ central memory T cells in the placebo group [4%], versus statistically significant increases in the active treatment groups [48% to 81%]. Similar trends were seen for β7+ effector memory T cells [placebo, 8%; PF-00547659, 84-138%] and β7+ naïve T cells [8%; 13-50%]. CCR9 gene expression had statistically significant up-regulation [p = 1.09e-06; false discovery rate < 0.1] with PF-00547659 treatment, and was associated with an increase in β7+ T cells. CONCLUSIONS Results of the OPERA study demonstrate positive pharmacology and dose-dependent changes in pharmacodynamic biomarker measurements in blood, including changes in cellular composition of lymphocytes and corresponding CCR9 gene expression changes.
Collapse
Affiliation(s)
- Mina Hassan-Zahraee
- Pfizer Inc., Cambridge, MA, USA,Corresponding Author: Mina Hassan-Zahraee, PhD, Pfizer Inc., 610 Main Street, Cambridge, MA 02139, USA. Tel.: 1 781-799-8694; fax: 973-660-8096;
| | | | | | | | | | | | | | | | | | | | | | - Li Xi
- Pfizer Inc., Cambridge, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Longmate W, DiPersio CM. Beyond adhesion: emerging roles for integrins in control of the tumor microenvironment. F1000Res 2017; 6:1612. [PMID: 29026524 PMCID: PMC5583736 DOI: 10.12688/f1000research.11877.1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2017] [Indexed: 12/21/2022] Open
Abstract
While integrins were originally discovered as cell adhesion receptors, recent studies have reinforced the concept that integrins have central roles in cancer that extend far beyond controlling cell adhesion and migration. Indeed, as transmembrane cell surface receptors that occupy a critical position at the interface of cellular and extracellular interactions and are capable of both "inside-out" and "outside-in" signaling, integrins are uniquely poised to regulate the cell's ability to promote, sense, and react to changes in the tumor microenvironment. Moreover, integrins are present on all cell types in the tumor microenvironment, and they have important roles in regulating intercellular communication. Decades of promising pre-clinical studies have implicated certain integrins as attractive therapeutic targets in the cancer clinic. Nevertheless, results of the few clinical trials that target integrins in cancer have thus far been disappointing. Importantly, these clinical failures likely reflect the emerging complexity of individual and combinatorial integrin function within both tumor cells and other cell types of the tumor microenvironment, together with a need to explore integrin-targeting agents not just as monotherapies but also as adjuvants to more conventional radiotherapies or chemotherapies. In this review, we will examine recent advances toward understanding how integrins regulate cancer progression, including their roles in intercellular communication and modulation of the tumor microenvironment. Additionally, we will discuss factors that underlie the limited efficacy of current efforts to target integrins in the cancer clinic as well as potential strategies to overcome these challenges.
Collapse
Affiliation(s)
- Whitney Longmate
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - C Michael DiPersio
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA.,Department of Surgery, Albany Medical College, Albany , New York, USA
| |
Collapse
|
21
|
Feng XB, Jiang J, Li M, Wang G, You JW, Zuo J. Role of intestinal flora imbalance in pathogenesis of pouchitis. ASIAN PAC J TROP MED 2016; 9:786-90. [PMID: 27569889 DOI: 10.1016/j.apjtm.2016.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To discuss the role of intestinal flora imbalance in the pathogenesis of pouchitis. METHODS The pouchitis rat model was established and the faeces sample and the mucous membrane sample were collected regularly, in which the bacterial nucleic acids were extracted for quantitative analysis of the intestinal flora in the samples through using the real-time quantitative PCR technique and high energy sequencing technology. RESULTS The disorder phenomenon of the intestinal flora appeared at the 7th day of the experiment, and the pouchitis was presented at the 21st day of the experiment. At the 31st day of the experiment, compared to control group and non-pouchitis group, the quantity of Bifidobacterium and the Lactobacillus of the pouchitis model rats in the mucous membrane sample and the faeces sample were significantly decreased (P < 0.05), and the Bacteroidetes, Faecalibacterium prausnitzii and XIV Clostridium leptum subgroup in the mucous membrane of pouchitis were significantly decreased (P < 0.05). The IV Clostridium coccoides group was the main flora in the mucous membrane of pouchitis, the bacterial diversity of non-pouchitis group and control group was significantly higher than that of the pouchitis group (P < 0.05). CONCLUSIONS The intestinal flora imbalance is one of the factors that cause the incidence of the pouchitis; this study provides a clue of the pathogenesis and treatment direction of the intestinal inflammatory disease.
Collapse
Affiliation(s)
- Xiao-Bo Feng
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu Province 210002, People's Republic of China
| | - Jun Jiang
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu Province 210002, People's Republic of China
| | - Min Li
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu Province 210002, People's Republic of China
| | - Gang Wang
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu Province 210002, People's Republic of China
| | - Jin-Wei You
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu Province 210002, People's Republic of China
| | - Jian Zuo
- State Key Laboratory of Infectious Disease Diagnosis and Treatment, First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| |
Collapse
|