1
|
Naaz A, Turnquist HR, Gorantla VS, Little SR. Drug delivery strategies for local immunomodulation in transplantation: Bridging the translational gap. Adv Drug Deliv Rev 2024; 213:115429. [PMID: 39142608 DOI: 10.1016/j.addr.2024.115429] [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: 02/15/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Drug delivery strategies for local immunomodulation hold tremendous promise compared to current clinical gold-standard systemic immunosuppression as they could improve the benefit to risk ratio of life-saving or life-enhancing transplants. Such strategies have facilitated prolonged graft survival in animal models at lower drug doses while minimizing off-target effects. Despite the promising outcomes in preclinical animal studies, progression of these strategies to clinical trials has faced challenges. A comprehensive understanding of the translational barriers is a critical first step towards clinical validation of effective immunomodulatory drug delivery protocols proven for safety and tolerability in pre-clinical animal models. This review overviews the current state-of-the-art in local immunomodulatory strategies for transplantation and outlines the key challenges hindering their clinical translation.
Collapse
Affiliation(s)
- Afsana Naaz
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, 15213, United States.
| | - Heth R Turnquist
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States.
| | - Vijay S Gorantla
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States; Departments of Surgery, Ophthalmology and Bioengineering, Wake Forest School of Medicine, Wake Forest Institute of Regenerative Medicine, Winston Salem, NC, 27101, United States.
| | - Steven R Little
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
2
|
Srinivasan S, Zhu C, McShan AC. Structure, function, and immunomodulation of the CD8 co-receptor. Front Immunol 2024; 15:1412513. [PMID: 39253084 PMCID: PMC11381289 DOI: 10.3389/fimmu.2024.1412513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Expressed on the surface of CD8+ T cells, the CD8 co-receptor is a key component of the T cells that contributes to antigen recognition, immune cell maturation, and immune cell signaling. While CD8 is widely recognized as a co-stimulatory molecule for conventional CD8+ αβ T cells, recent reports highlight its multifaceted role in both adaptive and innate immune responses. In this review, we discuss the utility of CD8 in relation to its immunomodulatory properties. We outline the unique structure and function of different CD8 domains (ectodomain, hinge, transmembrane, cytoplasmic tail) in the context of the distinct properties of CD8αα homodimers and CD8αβ heterodimers. We discuss CD8 features commonly used to construct chimeric antigen receptors for immunotherapy. We describe the molecular interactions of CD8 with classical MHC-I, non-classical MHCs, and Lck partners involved in T cell signaling. Engineered and naturally occurring CD8 mutations that alter immune responses are discussed. The applications of anti-CD8 monoclonal antibodies (mABs) that target CD8 are summarized. Finally, we examine the unique structure and function of several CD8/mAB complexes. Collectively, these findings reveal the promising immunomodulatory properties of CD8 and CD8 binding partners, not only to uncover basic immune system function, but to advance efforts towards translational research for targeted immunotherapy.
Collapse
Affiliation(s)
- Shreyaa Srinivasan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Cheng Zhu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Andrew C. McShan
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
3
|
Saowapa S, Polpichai N, Siladech P, Wannaphut C, Tanariyakul M, Wattanachayakul P, Lalitnithi P. Evaluating Kaposi Sarcoma in Kidney Transplant Patients: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e52527. [PMID: 38371002 PMCID: PMC10874301 DOI: 10.7759/cureus.52527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Kaposi's sarcoma (KS) is a malignancy that commonly appears as lesions on the skin or mucosal surfaces but can also develop in other organs. This cancer is usually caused by the human herpesvirus 8 (HHV-8), recently known as Kaposi's sarcoma-associated herpesvirus (KSHV). KS is rare in the general population but can develop in kidney transplant recipients with varying incidence due to immunocompromised status from immunosuppression. The main aim of the present systematic review was to identify the prevalence and treatment of KS in kidney transplant patients. PubMed, Cochrane Library, and Google Scholar databases were searched for studies until October 2023. Full-text studies with similar research objectives were included, while non-English articles, reviews, case reports, ongoing clinical trials, and studies evaluating KS in HIV patients or after other solid organ transplants were excluded. All studies were observational; therefore, methodological quality was assessed using the Newcastle-Ottawa Scale. The statistical analyses were performed with the Comprehensive Meta-Analysis (CMA) software (Biostat, Inc. Englewood, NJ). The pooled analysis from the 15 studies included showed that KS develops in 1.5% of kidney transplant recipients and is more prevalent in African (1.7%) and Middle Eastern (1.7%) recipients than in Western recipients (0.07%). KS was also significantly more prevalent among male recipients than female recipients (OR: 2.36; p < 0.0001). Additionally, cyclosporine-based immunosuppression accounts for most KS incidences (79.6%) compared to azathioprine-based immunosuppression (28.2%). Furthermore, reduction or withdrawal of immunosuppression alone resulted in 47.8% KS complete remissions. Post-kidney transplantation KS is more frequent among males and patients of Middle Eastern and African origin. However, the gender difference may be attributed to most patients undergoing kidney transplants being male. Therefore, if gender balance is considered in future studies, then the difference might be insignificant. Based on our results, we can concur that the mainstay treatment for post-transplant KS is reduction or withdrawal of immunosuppression. However, the patients should be closely monitored to avoid KS recurrence and kidney rejection. Furthermore, there is an increased risk for KS with the use of cyclosporine-based immunosuppression. However, this does not mean that the withdrawal of this immunosuppression agent might result in improved KS outcomes because the withdrawal of azathioprine with or without cyclosporine reduction has also led to improved outcomes.
Collapse
Affiliation(s)
- Sakditad Saowapa
- Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, USA
| | | | | | - Chalothorn Wannaphut
- Internal Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, USA
| | - Manasawee Tanariyakul
- Internal Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, USA
| | | | | |
Collapse
|
4
|
Tammisetti VS, Prasad SR, Dasyam N, Menias CO, Katabathina V. Immunosuppressive Therapy in Solid Organ Transplantation: Primer for Radiologists and Potential Complications. Radiol Clin North Am 2023; 61:913-932. [PMID: 37495297 DOI: 10.1016/j.rcl.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
The availability of effective immunosuppressive medication is primarily responsible for the dramatic improvement in long-term graft survival rates after solid organ transplantation. The commonly used drugs include monoclonal/polyclonal antibodies, corticosteroids, calcineurin inhibitors (cyclosporine and tacrolimus), antimetabolites, mammalian target of rapamycin, and many novel drugs. Prolonged immunosuppression is accompanied by several well-described potentially life-threatening complications. In addition to drug-related side effects, recipients of solid organs are unavoidably at a higher risk for infections and malignancies. Select infections and malignancies in solid organ transplant patients have distinctive imaging findings, and radiologists play a crucial role in the timely diagnosis and management of these conditions.
Collapse
Affiliation(s)
| | - Srinivasa R Prasad
- Department of Radiology, University of Texas M. D. Anderson Cancer Center
| | - Navya Dasyam
- Department of Radiology, University of Pittsburgh Medical Center
| | | | | |
Collapse
|
5
|
Lee J, Yang AWJ, Chung LIY, Yu J, Lee Y, Kim HS, Shin HJ, Choi YG, Bharat A, Chae YK. A Comprehensive Landscape of De Novo Malignancy After Double Lung Transplantation. Transpl Int 2023; 36:11552. [PMID: 37663524 PMCID: PMC10468575 DOI: 10.3389/ti.2023.11552] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023]
Abstract
Although the association between post-transplant malignancy (PTM) and immunosuppressive therapy after organ transplantation has been studied, an integrated review of PTM after lung transplantation is lacking. We investigated the incidence and types of de novo PTM and its impact on survival following double lung transplantation (DLT). The incidence and type of PTM as well as the annual and cumulative risks of each malignancy after DLT were analyzed. The overall survival (OS) of recipients with or without PTM was compared by the Kaplan-Meier survival method and landmark analysis. There were 5,629 cases (23.52%) with 27 types of PTMs and incidences and OS varied according to the types of PTMs. The recipients with PTM showed a significantly longer OS than those without PTM (p < 0.001). However, while the recipients with PTM showed significantly better OS at 3, and 5 years (p < 0.001, p = 0.007), it was worse at the 10-year landmark time (p = 0.013). And the single PTM group showed a worse OS rate than the multiple PTM group (p < 0.001). This comprehensive report on PTM following DLT can help understand the risks and timing of PTM to improve the implementation of screening and treatment.
Collapse
Affiliation(s)
- Jeeyeon Lee
- Department of Surgery, School of Medicine, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, Republic of Korea
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| | - Andrew Won Jun Yang
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| | - Liam Il-Young Chung
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| | - Jisang Yu
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| | - Yunjoo Lee
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| | - Hye Sung Kim
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| | - Hyun Joon Shin
- Division of Cardiology, Department of Medicine, Lemuel Shattuck Hospital, Massachusetts Department of Public Health, Jamaica Plain, MA, United States
| | - Young-Geun Choi
- Department of Mathematics Education, Sungkyunkwan University, Seoul, Republic of Korea
| | - Ankit Bharat
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| | - Young Kwang Chae
- Department of Internal Medicine, Northwestern Memorial Hospital, Chicago, IL, United States
| |
Collapse
|
6
|
Carrara SC, Harwardt J, Grzeschik J, Hock B, Kolmar H. TriTECM: A tetrafunctional T-cell engaging antibody with built-in risk mitigation of cytokine release syndrome. Front Immunol 2022; 13:1051875. [DOI: 10.3389/fimmu.2022.1051875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/21/2022] [Indexed: 11/12/2022] Open
Abstract
Harnessing the innate power of T cells for therapeutic benefit has seen many shortcomings due to cytotoxicity in the past, but still remains a very attractive mechanism of action for immune-modulating biotherapeutics. With the intent of expanding the therapeutic window for T-cell targeting biotherapeutics, we present an attenuated trispecific T-cell engager (TCE) combined with an anti- interleukin 6 receptor (IL-6R) binding moiety in order to modulate cytokine activity (TriTECM). Overshooting cytokine release, culminating in cytokine release syndrome (CRS), is one of the severest adverse effects observed with T-cell immunotherapies, where the IL-6/IL-6R axis is known to play a pivotal role. By targeting two tumour-associated antigens, epidermal growth factor receptor (EGFR) and programmed death ligand 1 (PD-L1), simultaneously with a bispecific two-in-one antibody, high tumour selectivity together with checkpoint inhibition was achieved. We generated tetrafunctional molecules that contained additional CD3- and IL-6R-binding modules. Ligand competition for both PD-L1 and IL-6R as well as inhibition of both EGF- and IL-6-mediated signalling pathways was observed. Furthermore, TriTECM molecules were able to activate T cells and trigger T-cell-mediated cytotoxicity through CD3-binding in an attenuated fashion. A decrease in pro-inflammatory cytokine interferon γ (IFNγ) after T-cell activation was observed for the TriTECM molecules compared to their respective controls lacking IL-6R binding, hinting at a successful attenuation and potential modulation via IL-6R. As IL-6 is a key player in cytokine release syndrome as well as being implicated in enhancing tumour progression, such molecule designs could reduce side effects and cytotoxicity observed with previous TCEs and widen their therapeutic windows.
Collapse
|
7
|
Sen T, Thummer RP. The Impact of Human Microbiotas in Hematopoietic Stem Cell and Organ Transplantation. Front Immunol 2022; 13:932228. [PMID: 35874759 PMCID: PMC9300833 DOI: 10.3389/fimmu.2022.932228] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
The human microbiota heavily influences most vital aspects of human physiology including organ transplantation outcomes and transplant rejection risk. A variety of organ transplantation scenarios such as lung and heart transplantation as well as hematopoietic stem cell transplantation is heavily influenced by the human microbiotas. The human microbiota refers to a rich, diverse, and complex ecosystem of bacteria, fungi, archaea, helminths, protozoans, parasites, and viruses. Research accumulating over the past decade has established the existence of complex cross-species, cross-kingdom interactions between the residents of the various human microbiotas and the human body. Since the gut microbiota is the densest, most popular, and most studied human microbiota, the impact of other human microbiotas such as the oral, lung, urinary, and genital microbiotas is often overshadowed. However, these microbiotas also provide critical and unique insights pertaining to transplantation success, rejection risk, and overall host health, across multiple different transplantation scenarios. Organ transplantation as well as the pre-, peri-, and post-transplant pharmacological regimens patients undergo is known to adversely impact the microbiotas, thereby increasing the risk of adverse patient outcomes. Over the past decade, holistic approaches to post-transplant patient care such as the administration of clinical and dietary interventions aiming at restoring deranged microbiota community structures have been gaining momentum. Examples of these include prebiotic and probiotic administration, fecal microbial transplantation, and bacteriophage-mediated multidrug-resistant bacterial decolonization. This review will discuss these perspectives and explore the role of different human microbiotas in the context of various transplantation scenarios.
Collapse
Affiliation(s)
| | - Rajkumar P. Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
8
|
Konda P, Golamari R, Eisen HJ. Novel Immunosuppression in Solid Organ Transplantation. Handb Exp Pharmacol 2022; 272:267-285. [PMID: 35318509 DOI: 10.1007/164_2021_569] [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] [Indexed: 06/14/2023]
Abstract
Solid organ transplantation and survival has improved tremendously in the last few decades, much of the success has been attributed to the advancements in immunosuppression. While steroids are being replaced and much of the immunosuppressive strategies focus on steroid free regimens, novel agents have introduced in the induction, maintenance, and treatment of acute rejection phase. MTOR inhibitors have helped with the renal sparing side effect from the calcineurin inhibitors, newer agents such as rituximab have decreased the incidence of donor-specific antibodies which led to decreased incidence of acute rejection reactions. In this chapter we discuss the newer therapies directed specifically for solid organ transplantation.
Collapse
Affiliation(s)
- Prasad Konda
- Heart and Vascular Institute, Pennsylvania State University/Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Reshma Golamari
- Department of Hospital Medicine, Pennsylvania State University/Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Howard J Eisen
- Heart and Vascular Institute, Pennsylvania State University/Milton S. Hershey Medical Center, Hershey, PA, USA.
| |
Collapse
|
9
|
Kaleta B. Osteopontin and Transplantation: Where Are We Now? Arch Immunol Ther Exp (Warsz) 2021; 69:15. [PMID: 34019147 PMCID: PMC8139897 DOI: 10.1007/s00005-021-00617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/12/2021] [Indexed: 11/26/2022]
Abstract
Organ transplantation represents the optimal therapeutic tool for patients with end-stage organ failure. Hematopoietic stem cell transplantation (HSCT) is likewise an effective therapy for a wide range of malignant and non-malignant diseases. Better understanding of transplantation immunology and the use of multi-modal immunosuppression protocols, can decrease the risk of graft failure and graft-versus-host disease (GVHD) after HSCT. Nevertheless, a major challenge of modern transplantology still seems to be finding non-invasive biomarkers for recipients selection, monitoring of allograft function, and diagnosis of rejection. Since proinflammatory cytokine osteopontin (OPN) is closely involved in regulating both adaptive and innate immune responses, as well as the pathogenesis of inflammatory and autoimmune diseases, it is likely to play an important role in organ and HSC transplantation. This review is to summarize recent advances in our knowledge about OPN function in the kidney, heart, liver, lung, and HSC transplantation. Most studies found that elevated OPN is associated with poorer graft function in kidney, heart, liver and lung recipients. Moreover, some reports suggested that this protein can play role in GVHD pathogenesis. However, due to relatively small number of similar studies, as well as some inconclusive results, future investigation in this field is needed to verify if OPN can serve as a biomarker of organ and HSC transplantation. The knowledge about such markers will promote our understanding of the mechanisms underlying graft dysfunction and posttransplant mortality. In addition, such knowledge may be helpful in the development of new treatment strategies and identification of recipients with increased risk of allograft failure.
Collapse
Affiliation(s)
- Beata Kaleta
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59 St., 02-006, Warsaw, Poland.
| |
Collapse
|
10
|
Horen SR, Lopez J, Dorafshar AH. Facial Transplantation. Facial Plast Surg 2021; 37:528-535. [PMID: 33831957 DOI: 10.1055/s-0041-1723766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Facial transplantation represents a unique surgical solution for challenging facial injury patterns in which conservative reconstructive techniques fail to provide a satisfactory functional and aesthetic result. With advances in the field of vascularized composite allotransplantation over the past 15 years, more than 40 of these procedures have been performed worldwide with two recent reports of facial re-transplantation. In this article we discuss the multidisciplinary approach that is required for successful transplantation as well as the surgical techniques used and postoperative management. With ongoing research, recent technological innovation, and increased efforts to promote greater generalizability and transparency in this field, patients with these complex injuries will continue to see improvements in their treatment options, and thus quality of life.
Collapse
Affiliation(s)
- Sydney R Horen
- Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL
| | - Joseph Lopez
- Division of Plastic Surgery, Yale University, New Haven, Connecticut
| | - Amir H Dorafshar
- Division of Plastic and Reconstructive Surgery, Rush University Medical Center, Chicago, IL
| |
Collapse
|
11
|
Ferreira-Silva M, Faria-Silva C, Baptista PV, Fernandes E, Fernandes AR, Corvo ML. Drug delivery nanosystems targeted to hepatic ischemia and reperfusion injury. Drug Deliv Transl Res 2021; 11:397-410. [PMID: 33660214 DOI: 10.1007/s13346-021-00915-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Hepatic ischemia and reperfusion injury (IRI) is an acute inflammatory process that results from surgical interventions, such as liver resection surgery or transplantation, or hemorrhagic shock. This pathology has become a severe clinical issue, due to the increasing incidence of hepatic cancer and the high number of liver transplants. So far, an effective treatment has not been implemented in the clinic. Despite its importance, hepatic IRI has not attracted much interest as an inflammatory disease, and only a few reviews addressed it from a therapeutic perspective with drug delivery nanosystems. In the last decades, drug delivery nanosystems have proved to be a major asset in therapy because of their ability to optimize drug delivery, either by passive or active targeting. Passive targeting is achieved through the enhanced permeability and retention (EPR) effect, a main feature in inflammation that allows the accumulation of the nanocarriers in inflammation sites, enabling a higher efficacy of treatment than conventional therapies. These systems also can be actively targeted to specific compounds, such as inflammatory markers and overexpressed receptors in immune system intermediaries, allowing an even more specialized therapy that have already showed encouraging results. In this manuscript, we review drug delivery nanosystems designed for hepatic IRI treatment, addressing their current state in clinical trials, discussing the main hurdles that hinder their successful translation to the market and providing some suggestions that could potentially advance their clinical translation.
Collapse
Affiliation(s)
- Margarida Ferreira-Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Catarina Faria-Silva
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Pedro Viana Baptista
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516, Caparica, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Alexandra Ramos Fernandes
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516, Caparica, Portugal
| | - Maria Luísa Corvo
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.
| |
Collapse
|
12
|
Kuwabara R, Hu S, Smink AM, Orive G, Lakey JRT, de Vos P. Applying Immunomodulation to Promote Longevity of Immunoisolated Pancreatic Islet Grafts. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:129-140. [PMID: 33397201 DOI: 10.1089/ten.teb.2020.0326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Islet transplantation is a promising therapy for insulin-dependent diabetes, but large-scale application is hampered by the lack of a consistent source of insulin-producing cells and need for lifelong administration of immunosuppressive drugs, which are associated with severe side effects. To avoid chronic immunosuppression, islet grafts can be enveloped in immunoisolating polymeric membranes. These immunoisolating polymeric membranes protect islet grafts from cell-mediated rejection while allowing diffusion of oxygen, nutrients, and insulin. Although clinical trials have shown the safety and feasibility of encapsulated islets to control glucose homeostasis, the strategy does up till now not support long-term graft survival. This partly can be explained by a significant loss of insulin-producing cells in the immediate period after implantation. The loss can be prevented by combining immunoisolation with immunomodulation, such as combined administration of immunomodulating cytokines or coencapsulation of immunomodulating cell types such as regulatory T cells, mesenchymal stem cells, or Sertoli cells. Also, administration of specific antibodies or apoptotic donor leucocytes is considered to create a tolerant microenvironment around immunoisolated grafts. In this review, we describe the outcomes and limitations of these approaches, as well as the recent progress in immunoisolating devices. Impact statement Immunoisolation by enveloping islets in semipermeable membranes allows for successful transplantation of islet grafts in the absence of chronic immunosuppression, but the duration of graft survival is still not permanent. The reasons for long-term final graft failure is not fully understood, but combining immunoisolation with immunomodulation of tissues or host immune system has been proposed to enhance the longevity of grafts. This article reviews the recent progress and challenges of immunoisolation, as well as the benefits and feasibility of combining encapsulation approaches with immunomodulation to promote longevity of encapsulated grafts.
Collapse
Affiliation(s)
- Rei Kuwabara
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shuxian Hu
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexandra M Smink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Jonathan R T Lakey
- Department of Surgery and Biomedical Engineering, University of California Irvine, Irvine, California, USA
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Anugwom CM, Parekh JR, Hwang C, MacConmara M, Lee WM, Leventhal TM. Comparison of Clinical Outcomes of Induction Regimens in Patients Undergoing Liver Transplantation for Acute Liver Failure. Liver Transpl 2021; 27:27-33. [PMID: 32578297 DOI: 10.1002/lt.25832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023]
Abstract
Spontaneous survival rates in acute liver failure (ALF) are vastly improved by liver transplantation (LT). However, the value of induction agents beyond steroids continues to be debated. To understand the potential benefit of different induction regimens in the ALF population, we compared overall survival of recipients undergoing LT in the United States for ALF. Using the Scientific Registry of Transplant Recipients, we assessed the impact of induction immunosuppression (IS) in a cohort of 3754 first-time LT recipients with a diagnosis of ALF from 2002 to 2018. Induction IS therapy was grouped into steroid-only induction, use of antithymocyte globulin (ATG), or interleukin 2 receptor antibody. Other regimens were excluded from analysis. Survival analysis was estimated via Cox proportional hazards models and expressed as hazard ratios (HRs). In LT for ALF, the use of induction agents beyond steroids is increasingly frequent over the last 2 decades. The use of ATG is associated with worse overall survival, even after adjusting for donor and recipient factors, with HR of 1.24 (95% confidence interval, 1.00-1.53; P = 0.05). An elevated serum creatinine, recipient and donor age, and Black ethnicity, were all associated with reduced survival, whereas maintenance IS with calcineurin inhibitors (CNIs) was associated with improved survival. Although adjunct induction therapy has become more common, our analysis shows that compared with a steroid-only induction regimen, the addition of ATG is associated with worse overall survival after LT for ALF. CNI maintenance was highly protective, suggesting that an IS strategy focusing on corticosteroid-only induction followed by CNI maintenance may offer the best overall survival rate.
Collapse
Affiliation(s)
- Chimaobi M Anugwom
- Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical Center, Minneapolis, MN
| | - Justin R Parekh
- Department of Surgery, University of California, San Diego, San Diego, CA
| | - Christine Hwang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Malcolm MacConmara
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - William M Lee
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Thomas M Leventhal
- Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical Center, Minneapolis, MN
| |
Collapse
|
14
|
Pilch NA, Bowman LJ, Taber DJ. Immunosuppression trends in solid organ transplantation: The future of individualization, monitoring, and management. Pharmacotherapy 2020; 41:119-131. [PMID: 33131123 DOI: 10.1002/phar.2481] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/07/2020] [Accepted: 08/09/2020] [Indexed: 12/20/2022]
Abstract
Immunosuppression regimens used in solid organ transplant have evolved significantly over the past 70 years in the United States. Early immunosuppression and targets for allograft success were measured by incidence and severity of allograft rejection and 1-year patient survival. The limited number of agents, infancy of human leukocyte antigen (HLA) matching techniques and lack of understanding of immunoreactivity limited the early development of effective regimens. The 1980s and 1990s saw incredible advancements in these areas, with acute rejection rates halving in a short span of time. However, the constant struggle to achieve the optimal balance between under- and overimmunosuppression is weaved throughout the history of transplant immunosuppression. The aim of this paper is to discuss the different eras of immunosuppression and highlight the important milestones that were achieved while also discussing this in the context of rational agent selection and regimen design. This discussion sets the stage for how we can achieve optimal long-term outcomes during the next era of immunosuppression, which will move from universal protocols to patient-specific optimization.
Collapse
Affiliation(s)
- Nicole A Pilch
- Department of Pharmacy Practice and Outcomes Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lyndsey J Bowman
- Department of Pharmacy, Tampa General Hospital, Tampa, Florida, USA
| | - David J Taber
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Pharmacy Services, Ralph H. Johnson VAMC, Charleston, South Carolina, USA
| |
Collapse
|
15
|
Rapid Affinity Maturation of Novel Anti-PD-L1 Antibodies by a Fast Drop of the Antigen Concentration and FACS Selection of Yeast Libraries. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6051870. [PMID: 31976323 PMCID: PMC6959147 DOI: 10.1155/2019/6051870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022]
Abstract
The affinity engineering is a key step to increase the efficacy of therapeutic monoclonal antibodies and yeast surface display is the most widely used and powerful affinity maturation approach, achieving picomolar binding affinities. In this study, we provide an optimization of the yeast surface display methodology, applied to the generation of potentially therapeutic high affinity antibodies targeting the immune checkpoint PD-L1. In this approach, we coupled a 10-cycle error-prone mutagenesis of heavy chain complementarity determining region 3 of an anti‐PD-L1 scFv, previously identified by phage display, with high-throughput sequencing, to generate scFv-yeast libraries with high mutant frequency and diversity. In addition, we set up a novel, faster and effective selection scheme by fluorescence-activated cell sorting, based on a fast drop of the antigen concentration between the first and the last selection cycles, unlike the gradual decrease typical of current selection protocols. In this way we isolated 6 enriched mutated scFv-yeast clones overall, showing an affinity improvement for soluble PD-L1 protein compared to the parental scFv. As a proof of the potency of the novel approach, we confirmed that the antibodies converted from all the mutated scFvs retained the affinity improvement. Remarkably, the best PD-L1 binder among them also bound with a higher affinity to PD-L1 expressed in its native conformation on human-activated lymphocytes, and it was able to stimulate lymphocyte proliferation in vitro more efficiently than its parental antibody. This optimized technology, besides the identification of a new potential checkpoint inhibitor, provides a tool for the quick isolation of high affinity binders.
Collapse
|
16
|
Albuquerque I, Monteiro AR, Soares N, Ferreira S. Encephalopathy in a kidney transplant recipient. BMJ Case Rep 2019; 12:12/9/e231077. [PMID: 31494589 DOI: 10.1136/bcr-2019-231077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
A 60-year-old man presented several times to the emergency department due to confusion and behavioral changes. He was a kidney transplant recipient dependent on hemodialysis due, presumably, to chronic nephropathy of the transplanted kidney, and was not under any immunosuppressive therapy. He was admitted to the hospital ward due to elevation of C reactive protein and severe proteinuria, leukocyturia and erythrocyturia. The alterations found in the spot urine examination were suggestive of nephritic syndrome, consistent with chronic nephropathy of the transplanted kidney. The neurologic deterioration, however, remained unexplained. CT of the brain and cerebrospinal fluid examination were unremarkable. Infection, auto-immune disease and malignancy were excluded. Corticoid therapy was started for rejection nephropathy. The patient improved dramatically and ultimately the transplanted kidney was removed. Chronic nephropathy of the transplanted kidney was confirmed histologically and the patient remained clinically asymptomatic, without corticoid therapy.
Collapse
Affiliation(s)
- Inês Albuquerque
- Internal Medicine, Centro Hospitalar de Sao Joao EPE, Porto, Portugal
| | - Ana Raquel Monteiro
- Instituto Portugues de Oncologia de Coimbra Francisco Gentil EPE, Coimbra, Portugal
| | - Neuza Soares
- Internal Medicine, Centro Hospitalar de Sao Joao EPE, Porto, Portugal
| | - Susana Ferreira
- Internal Medicine, Centro Hospitalar de Sao Joao EPE, Porto, Portugal
| |
Collapse
|
17
|
Cajanding R. Immunosuppression following organ transplantation. Part 1: mechanisms and immunosuppressive agents. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2018; 27:920-927. [PMID: 30187798 DOI: 10.12968/bjon.2018.27.16.920] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Solid organ transplantation has revolutionised medical care by providing a definitive cure for a wide spectrum of end-stage medical conditions. This treatment, however, does not come without complications and poses the risks of rejection, life-threatening infection, malignancies and recurrent organ failure, with significant impacts on patient outcomes. One of the major challenges involved in optimising post-transplant outcomes is managing the immune system's response to the transplanted graft and preventing organ rejection. This is mainly accomplished through the use of immunosuppressant agents, which have become a mainstay of treatment for a majority of post-transplant patients. This article, the first of two parts, discusses the concept of immunosuppression and its importance in the care of patients who have received an organ transplant. It focuses on the pathophysiologic mechanisms involved in transplant rejection and discusses the pharmacologic aspects of immunosuppression and its implications for patient care.
Collapse
Affiliation(s)
- Ruff Cajanding
- Staff Nurse, Liver Intensive Therapy Unit, Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London
| |
Collapse
|
18
|
Lim S, Kirkiles-Smith NC, Pober JS, Bothwell ALM, Choi JM. Regulation of human T cell responses by dNP2-ctCTLA-4 inhibits human skin and microvessel graft rejection. Biomaterials 2018; 183:128-138. [PMID: 30165256 DOI: 10.1016/j.biomaterials.2018.08.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/14/2018] [Accepted: 08/20/2018] [Indexed: 12/30/2022]
Abstract
Manipulation of human T cell functioning by delivery of macromolecules such as DNA, RNA, or protein is limited, unless the human T cells have been stimulated or electropermeabilized. To achieve successful adaptation and survival of a grafted organ, the alloreactive T cells that induce graft rejection must be regulated. Corticosteroids, calcineurin inhibitors, and mTOR inhibitors, which are systemic immunosuppressants, are currently used for transplantation, with significant side effects. In this study, we demonstrated that a cell-permeable peptide (CPP), dNP2, could efficiently deliver proteins into human CD4 and CD8 T cells. We confirmed regulatory functioning of the cytoplasmic domain of CTLA-4 conjugated with dNP2 (dNP2-ctCTLA-4) in human T cell activation, proliferation, and chemokine receptor expression. We utilized a human skin allograft system in SCID/beige mice to examine whether dNP2-ctCTLA-4 could inhibit allograft rejection by controlling T cell responses. The grafted skin tissue inflammation, allogeneic T cell infiltration, and blood cytokine level was markedly reduced by dNP2-ctCTLA-4, resulting in successful transplantation. In addition, it also inhibited T cell alloresponses against microvessels formed form Bcl-2-transduced human umbilical vein endothelial cells implanted into Balb/c Rag1-/-/IL-2Rγ-/- double knockout (DKO) mice, assessed as reduced T cell infiltration and granzyme B expression. These results collectively suggest that dNP2 peptide conjugation offers a valuable tool for delivering macromolecules like proteins into human T cells, and dNP2-ctCTLA-4 is a novel agent that shows potential in controlling human T cell responses to allow successful adaptation of grafted tissues.
Collapse
Affiliation(s)
- Sangho Lim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea
| | - Nancy C Kirkiles-Smith
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, Korea; Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Korea.
| |
Collapse
|
19
|
Cyclophosphamide for Refractory Acute Cellular Rejection After Lung Transplantation. Transplant Direct 2018; 4:e350. [PMID: 29796421 PMCID: PMC5959344 DOI: 10.1097/txd.0000000000000790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/28/2018] [Indexed: 11/30/2022] Open
Abstract
Background Acute cellular rejection (ACR) is a major risk factor for chronic lung allograft dysfunction after lung transplantation. Acute cellular rejection can persist or recur despite augmentation of immunosuppression by conventional methods. There are limited therapeutic options in treating these recurrent and refractory ACRs. We describe our experience with cyclophosphamide therapy for recurrent and refractory ACR in lung transplant recipients. Methods Six consecutive patients who were treated with cyclophosphamide for recurrent or refractory ACR were included in the series. The primary outcome measures were improvement in ACR score and forced expiratory volume at 1 second. Secondary outcome measures included adverse drug events including bone marrow suppression, gastrointestinal side effects, and infections. Results Five of the 6 patients treated demonstrated complete resolution of ACR on follow-up biopsies. Acute cellular rejection score improved after cyclophosphamide treatment (P = 0.03). None of the patients had high grade (≥A3) ACR in the 3 months after cyclophosphamide administration. Cyclophosphamide had no effect on forced expiratory volume at 1 second trend or bronchiolitis obliterans score. All patients tolerated cyclophosphamide with minor gastrointestinal side effects, mild bone marrow suppression, and nonfatal infections that were amenable to treatment. Conclusions Cyclophosphamide therapy is an option in treating recurrent and refractory ACR in patients who have failed conventional treatments. Cyclophosphamide is tolerated well without serious adverse drug events (ADE).
Collapse
|
20
|
Protein A affinity chromatography of Chinese hamster ovary (CHO) cell culture broths containing biopharmaceutical monoclonal antibody (mAb): Experiments and mechanistic transport, binding and equilibrium modeling. J Chromatogr B Analyt Technol Biomed Life Sci 2018. [DOI: 10.1016/j.jchromb.2018.02.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
21
|
Kang K, Chung J, Yang J, Kim H. Current Perspectives on Emerging CAR-Treg Cell Therapy: Based on Treg Cell Therapy in Clinical Trials and the Recent Approval of CAR-T Cell Therapy. KOREAN JOURNAL OF TRANSPLANTATION 2017. [DOI: 10.4285/jkstn.2017.31.4.157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Koeun Kang
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jaeseok Yang
- Transplantation Center, Seoul, Korea
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyori Kim
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| |
Collapse
|
22
|
Evaluation of Immunosuppressive Therapy Use for Tracheal Transplantation with Trachea-Mimetic Bellows Scaffolds in a Rabbit Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5205476. [PMID: 29226141 PMCID: PMC5684528 DOI: 10.1155/2017/5205476] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/10/2017] [Indexed: 11/28/2022]
Abstract
The objective of this study was to evaluate the use of immunosuppressive therapy with high-dose cyclosporine, high-dose azathioprine, and a combination of low-dose cyclosporine and azathioprine after tracheal reconstruction by using a trachea-mimetic graft of polycaprolactone (PCL) bellows-type scaffold in a rabbit model. Twenty-four healthy New Zealand white rabbits were used in the study. All underwent circumferential tracheal replacement using tissue-engineered tracheal graft, prepared from PCL bellows scaffold reinforced with silicone ring, collagen hydrogel, and human turbinate mesenchymal stromal cell (hTMSC) sheets. The control group (Group 1) received no medication. The three experimental groups were given daily cyclosporine intramuscular doses of 10 mg/kg (Group 2), azathioprine oral doses of 5 mg/kg (Group 3), and azathioprine oral doses of 2.5 mg/kg plus cyclosporine intramuscular doses of 5 mg/kg (Group 4) for 4 weeks or until death. Group 1 had longer survival times compared to Group 2 or Group 3. Each group except for Group 1 experienced decreases in amount of nutrition and weight loss. In addition, compared with the other groups, Group 2 had significantly increased serum interleukin-2 and interferon-γ levels 7 days after transplantation. The results of this study showed that the administration of cyclosporine and/or azathioprine after tracheal transplantation had no beneficial effects. Furthermore, the administration of cyclosporine had side effects, including extreme weight loss, respiratory distress, and diarrhea. Therefore, cyclosporine and azathioprine avoidance may be recommended for tracheal reconstruction using a native trachea-mimetic graft of PCL bellows-type scaffold in a rabbit model.
Collapse
|
23
|
Duff CE, Simmonds MJ. Genetic predictors of long-term graft function in kidney and pancreas transplant patients. Brief Funct Genomics 2017; 16:228-237. [PMID: 28110269 DOI: 10.1093/bfgp/elw039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Kidney and pancreas transplantation have helped transform the lives of people with end-stage renal failure and individuals with type 1 diabetes who have poor glycaemic control/severe secondary complications, respectively. Despite an improvement in immunosuppressive regimes, operative techniques and decreased initial rejection rates, there has been little improvement in long-term graft survival rates over the past decade. Whilst limited progress has been made in establishing clinical markers of graft function, several genetic markers of long-term graft function have been identified. These genetic markers have the potential to (i) assist in selecting marginal donor organs for transplantation, (ii) provide better understanding of the mechanisms behind graft loss enabling identification of new, or repurposing, current treatments to extend graft function and (iii) provide a window of opportunity to identify and treat individuals before graft failure has occurred. This review will discuss the different genetic variants screened for a role in predicting transplant longevity, examine their findings and limitations and introduce where the future of genetic research within the transplantation field lies.
Collapse
|
24
|
Pinho A, Gouveia M, Cardoso JC, Xavier MM, Vieira R, Alves R. Non-melanoma skin cancer in Portuguese kidney transplant recipients - incidence and risk factors. An Bras Dermatol 2017; 91:455-62. [PMID: 27579740 PMCID: PMC4999103 DOI: 10.1590/abd1806-4841.20164891] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/09/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cancer is currently among the three leading causes of death after solid organ transplantation and its incidence is increasing. Non-melanoma skin cancer - squamous cell carcinoma and basal cell carcinoma - is the most common malignancy found in kidney transplant recipients (KTRs). The incidence of non-melanoma skin cancer in KTRs has not been extensively studied in Portugal. OBJECTIVES To determine the incidence of non-melanoma skin cancer in KTRs from the largest Portuguese kidney transplant unit; and to study risk factors for non-melanoma skin cancer. METHODS Retrospective analysis of clinical records of KTRs referred for the first time for a dermatology consultation between 2004 and 2013. A case-control study was performed on KTRs with and without non-melanoma skin cancer. RESULTS We included 288 KTRs with a median age at transplantation of 47 years, a male gender predominance (66%) and a median transplant duration of 3.67 years. One fourth (n=71) of KTRs developed 131 non-melanoma skin cancers, including 69 (53%) squamous cell carcinomas and 62 (47%) basal cell carcinomas (ratio squamous cell carcinoma: basal cell carcinoma 1.11), with a mean of 1.85 neoplasms per patient. Forty percent of invasive squamous cell carcinomas involved at least two clinical or histological high-risk features. The following factors were associated with a higher risk of non-melanoma skin cancer: an older age at transplantation and at the first consultation, a longer transplant duration and the presence of actinic keratosis. KTRs treated with azathioprine were 2.85 times more likely to develop non-melanoma skin cancer (p=0.01). CONCLUSION Non-melanoma skin cancer was a common reason for dermatology consultation in Portuguese KTRs. It is imperative for KTRs to have access to specialized dermatology consultation for early referral and treatment of skin malignancies.
Collapse
Affiliation(s)
- André Pinho
- Centro Hospitalar e Universitário de Coimbra - Coimbra, Portugal
| | - Miguel Gouveia
- Centro Hospitalar e Universitário de Coimbra - Coimbra, Portugal
| | | | | | - Ricardo Vieira
- Centro Hospitalar e Universitário de Coimbra - Coimbra, Portugal
| | - Rui Alves
- Centro Hospitalar e Universitário de Coimbra - Coimbra, Portugal
| |
Collapse
|
25
|
Picarda E, Bézie S, Boucault L, Autrusseau E, Kilens S, Meistermann D, Martinet B, Daguin V, Donnart A, Charpentier E, David L, Anegon I, Guillonneau C. Transient antibody targeting of CD45RC induces transplant tolerance and potent antigen-specific regulatory T cells. JCI Insight 2017; 2:e90088. [PMID: 28194440 DOI: 10.1172/jci.insight.90088] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Rat and human CD4+ and CD8+ Tregs expressing low levels of CD45RC have strong immunoregulatory properties. We describe here that human CD45 isoforms are nonredundant and identify distinct subsets of cells. We show that CD45RC is not expressed by CD4+ and CD8+ Foxp3+ Tregs, while CD45RA/RB/RO are. Transient administration of a monoclonal antibody (mAb) targeting CD45RC in a rat cardiac allotransplantation model induced transplant tolerance associated with inhibition of allogeneic humoral responses but maintained primary and memory responses against cognate antigens. Anti-CD45RC mAb induced rapid death of CD45RChigh T cells through intrinsic cell signaling but preserved and potentiated CD4+ and CD8+ CD45RClow/- Tregs, which are able to adoptively transfer donor-specific tolerance to grafted recipients. Anti-CD45RC treatment results in distinct transcriptional signature of CD4+ and CD8+ CD45RClow/- Tregs. Finally, we demonstrate that anti-human CD45RC treatment inhibited graft-versus-host disease (GVHD) in immune-humanized NSG mice. Thus, short-term anti-CD45RC is a potent therapeutic candidate to induce transplantation tolerance in human.
Collapse
Affiliation(s)
- Elodie Picarda
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Séverine Bézie
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Laetitia Boucault
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Elodie Autrusseau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Stéphanie Kilens
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Dimitri Meistermann
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Bernard Martinet
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Véronique Daguin
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Audrey Donnart
- INSERM UMR1087, CNRS UMR6291, Université de Nantes, l'institut du thorax, Nantes, France
| | - Eric Charpentier
- INSERM UMR1087, CNRS UMR6291, Université de Nantes, l'institut du thorax, Nantes, France
| | - Laurent David
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
26
|
Grabowska M, Kędzierska K, Michałek K, Słuczanowska-Głąbowska S, Grabowski M, Piasecka M, Kram A, Rotter I, Rył A, Laszczyńska M. Effects of an immunosuppressive treatment on the rat prostate. Drug Des Devel Ther 2016; 10:2899-2915. [PMID: 27672312 PMCID: PMC5026216 DOI: 10.2147/dddt.s111695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The aim of this study was to determine the influence of different combinations of immunosuppressive drugs on the morphology, ultrastructure, and expression of proliferating cell nuclear antigen and cytoskeleton proteins in the rat dorsolateral prostate. The studies were conducted on 48 male Wistar rats. The animals were divided into eight groups: a control group and seven experimental groups. For 6 months, the animals in the experimental groups were administered a combination of drugs including rapamycin (Rapa), cyclosporin A, tacrolimus (Tac), mycophenolate mofetil, and prednisone (Pred), according to the standard three-drug regimens for immunosuppressive therapy used in clinical practice. An evaluation of the morphology and ultrastructure was conducted, and a quantitative evaluation of the expression of proliferating cell nuclear antigen and desmin- and cytokeratin-positive cells with weak, moderate, and strong expression was performed. The combination of Rapa, Tac, and Pred caused the smallest morphological and ultrastructural changes in the rat prostate cells. In the case of rats whose treatment was switched to Rapa monotherapy, a decreased percentage of proliferating cells of both the glandular epithelium and the stroma was found. Decreases in body weight and changes in the expression of cytokeratin and desmin were observed in all the experimental rats. The combination of Rapa, Tac, and Pred would seem to be the most beneficial for patients who do not suffer from prostate diseases. Our results justify the use of inhibitors of the mammalian target of Rapa in the treatment of patients with prostate cancer. The changes in the expression of cytoskeleton proteins may be the result of direct adverse effects of the immunosuppressive drugs, which are studied in this article, on the structure and organization of intermediate filament proteins.
Collapse
Affiliation(s)
- Marta Grabowska
- Department of Histology and Developmental Biology, Pomeranian Medical University
| | - Karolina Kędzierska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University
| | - Katarzyna Michałek
- Department of Physiology, Cytobiology and Proteomics, West Pomeranian University of Technology
| | | | - Maciej Grabowski
- Department of Microbiology and Applied Biotechnology, West Pomeranian University of Technology
| | - Małgorzata Piasecka
- Department of Histology and Developmental Biology, Pomeranian Medical University
| | - Andrzej Kram
- Department of Pathology, West Pomeranian Oncology Center
| | - Iwona Rotter
- Department of Medical Rehabilitation, Pomeranian Medical University, Szczecin, Poland
| | - Aleksandra Rył
- Department of Histology and Developmental Biology, Pomeranian Medical University
| | - Maria Laszczyńska
- Department of Histology and Developmental Biology, Pomeranian Medical University
| |
Collapse
|
27
|
Yun JW, Kim YY, Ahn JH, Kang BC, Ku SY. Use of nonhuman primates for the development of bioengineered female reproductive organs. Tissue Eng Regen Med 2016; 13:323-334. [PMID: 30603414 DOI: 10.1007/s13770-016-9091-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/13/2015] [Accepted: 12/09/2015] [Indexed: 01/02/2023] Open
Abstract
Nonhuman primates (NHPs) have been widely used in reproductive biology, neuroscience, and drug development since a number of primate species are phylogenetically close to humans. In this review, we summarize the use of NHPs for nonclinical application in the reproductive system disorders including the loss or failure of an organ or tissue. Causes of infertility include congenital aplasia and acquired disorders of the reproductive organs. In addition, anti-cancer treatments can deplete ovarian follicles, leading to premature ovarian failure, infertility and long-term health risks. Along with a limited supply of human reproductive organs, anatomic/physiologic similarities to humans support the need for NHP models (New-World monkeys such as the common marmoset and Old-World monkeys such as cynomolgus and rhesus monkeys) to promote the advances in female infertility studies. For maintaining and executing animal studies using NHP, special protocols including animal care, anesthetic protocol, surgical technique, and immunosuppressive protocol are necessary. With a growing interest in the potential therapies such as endometrial tissue engineering, and ovary/follicle cryopreservation and grafting in Korea, this review can be useful in selecting appropriate animal models and can bridge between nonclinical studies and clinical applications by providing detailed information on the use of NHPs in the field of reproductive organ disorders.
Collapse
Affiliation(s)
- Jun-Won Yun
- 1Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Yoon Young Kim
- 2Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Hun Ahn
- 1Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,3Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Byeong-Cheol Kang
- 1Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.,3Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea.,4Designed Animal Research Center, Institute of GreenBio Science Technology, Seoul National University, Pyeongchang, Korea.,5Biomedical Center for Animal Resource and Development, N-BIO, Seoul National University, Seoul, Korea.,6Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| | - Seung-Yup Ku
- 2Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,7Graduate School of Translational Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Korea
| |
Collapse
|
28
|
Donati M, Paolino G, Muscardin L, Panetta C, Donati P. Resolution of Benign and Malignant Sebaceous Neoplasms, in a Renal Transplant Patient Treated With Everolimus. EXP CLIN TRANSPLANT 2015; 15:100-102. [PMID: 25924010 DOI: 10.6002/ect.2014.0208] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nonmelanoma skin cancers are the most common malignancies in transplant recipients under immunosuppression; nevertheless, appendage tumors also may appear. The onset of several cutaneous neoplasms in transplant patients can cause deterioration in quality of life of these patients. A 62-year-old white woman patient developed several malignant and benign sebaceous neoplasms during an immunosuppressive treatment for a renal transplant. The genetic study showed a mutation in MSH6-eson 1 (c116G>A), without mutations in MLH1 gene and MSH2. A final diagnosis of multiple sebaceous tumors in an immunosuppressed patient without Muir -Torre syndrome was made. The spreading of further cutaneous neoplasms led to a change in immunosuppression: namely, that clinicians suspended tacrolimus and add everolimus. After 2 months, all tumor lesions on the face and on the limbs have disappeared, and no further lesions occurred. Everolimus could represent a valid therapeutical treatment for transplant patients at high risk for cutaneous tumors. A genetic consult and a consequent study of the genetic profile should be performed on each of these patients, to avoid risks of recurrent cutaneous tumors and negative effects on the quality of life.
Collapse
Affiliation(s)
- Michele Donati
- From the Dermatopathological Laboratory "San Gallicano Institute of Rome"; the Via Elio Chianesi, 53, 00158, Roma, Italy
| | | | | | | | | |
Collapse
|
29
|
Denny JT, Burr AT, Balzer F, Tse JT, Denny JE, Chyu D. Methylene blue treatment for cytokine release syndrome-associated vasoplegia following a renal transplant with rATG infusion: A case report and literature review. Exp Ther Med 2015; 9:1915-1920. [PMID: 26136914 DOI: 10.3892/etm.2015.2349] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 03/03/2015] [Indexed: 12/29/2022] Open
Abstract
Rabbit anti-thymocyte globulin (rATG) is an infusion of polyclonal rabbit-derived antibodies against human thymocyte markers, which can be used to prevent and treat acute rejection following organ transplantation. However, the product monograph issued by the manufacturer (Sanofi Canada) reports that serious immune-mediated reactions have been observed following the use of rATG, consisting of anaphylaxis or severe cytokine release syndrome (CRS), which is a form of vasoplegic syndrome (VS), in which distributive shock occurs refractory to norepinephrine (NE) and vasopressin (VP). Severe infusion-associated reactions are consistent with CRS and can cause serious cardiac or respiratory problems, or in certain cases, mortality. CRS is a form of systemic inflammatory response syndrome (SIRS). In SIRS, the substantial activation of endothelial inducible nitric oxide synthase (iNOS) and smooth muscle guanylate cyclase (GC) is observed, which can produce severe hypotension that is unresponsive to conventional vasopressors. Methylene blue (MB) is a direct inhibitor of iNOS and GC and has been used as an effective treatment for VS following cardiothoracic surgery. In the present study, the successful use of MB as a rescue therapy for CRS in a patient receiving rATG following a renal transplant was reported. Following an uneventful cadaveric kidney transplant involving the intravenous (IV) administration of rATG for the induction of immunological tolerance, the patient became markedly hypotensive and tachycardic. The patient required high doses of VP and NE infusions. Following the protocol described for treating refractory VS in post-cardiac surgery patients, the decision was made to initiate the patient on an IV MB infusion. This treatment protocol was shown to improve the hemodynamic status of the patient, which enabled the withdrawal of vasopressors and suggests an important role for methylene blue in the management of refractory VS.
Collapse
Affiliation(s)
- John T Denny
- Department of Anesthesia, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Andrew T Burr
- Department of Anesthesia, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Fred Balzer
- Virtua Memorial Hospital, Department of Anesthesia, Mount Holly, NJ 08060, USA
| | - James T Tse
- Department of Anesthesia, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Julia E Denny
- Department of Anesthesia, New York University Medical Center, New York, NY 10016, USA
| | - Darrick Chyu
- Department of Anesthesia, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
30
|
Immunology of Transplant Protocols. CURRENT OTORHINOLARYNGOLOGY REPORTS 2014. [DOI: 10.1007/s40136-014-0057-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
31
|
Tmem time: memory T-cells in cardiac allograft vasculopathy : editorial to: "memory t cells mediate cardiac allograft vasculopathy and are inactivated by anti-OX40L monoclonal antibody" by Hao Wang et al. Cardiovasc Drugs Ther 2014; 28:111-2. [PMID: 24384980 PMCID: PMC3955131 DOI: 10.1007/s10557-013-6507-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
32
|
FcRn: from molecular interactions to regulation of IgG pharmacokinetics and functions. Curr Top Microbiol Immunol 2014; 382:249-72. [PMID: 25116104 DOI: 10.1007/978-3-319-07911-0_12] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neonatal Fc receptor, FcRn, is related to MHC class I with respect to its structure and association with β2microglobulin (β2m). However, by contrast with MHC class I molecules, FcRn does not bind to peptides, but interacts with the Fc portion of IgGs and belongs to the Fc receptor family. Unlike the 'classical' Fc receptors, however, the primary functions of FcRn include salvage of IgG (and albumin) from lysosomal degradation through the recycling and transcytosis of IgG within cells. The characteristic feature of FcRn is pH-dependent binding to IgG, with relatively strong binding at acidic pH (<6.5) and negligible binding at physiological pH (7.3-7.4). FcRn is expressed in many different cell types, and endothelial and hematopoietic cells are the dominant cell types involved in IgG homeostasis in vivo. FcRn also delivers IgG across cellular barriers to sites of pathogen encounter and consequently plays a role in protection against infections, in addition to regulating renal filtration and immune complex-mediated antigen presentation. Further, FcRn has been targeted to develop both IgGs with extended half-lives and FcRn inhibitors that can lower endogenous antibody levels. These approaches have implications for the development of longer lived therapeutics and the removal of pathogenic or deleterious antibodies.
Collapse
|
33
|
Pant C, Deshpande A, Larson A, O'Connor J, Rolston DDK, Sferra TJ. Diarrhea in solid-organ transplant recipients: a review of the evidence. Curr Med Res Opin 2013; 29:1315-28. [PMID: 23777312 DOI: 10.1185/03007995.2013.816278] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To provide a comprehensive review of the literature as it relates to diarrhea in solid organ transplant (SOT) recipients. In this article, we review the epidemiology, pathogenesis, clinical manifestations, diagnosis and management of diarrhea in SOT recipients and discuss recent advances and challenges. METHODS Two investigators conducted independent literature searches using PubMed, Web of Science, and Scopus until January 1st, 2013. All databases were searched using a combination of the terms diarrhea, solid organ transplant, SOT, transplant associated diarrhea, and transplant recipients. Articles that discussed diarrhea in SOT recipients were reviewed and relevant cross-references also read and evaluated for inclusion. Selection bias could be a possible limitation of the approach used in selecting or finding articles for this article. FINDINGS Post-transplant diarrhea is a common and distressing occurrence in patients, which can have significant deleterious effects on the clinical course and well-being of the organ recipient. A majority of cases are due to infectious and drug-related etiologies. However, various other etiologies including inflammatory bowel disease must be considered in the differential diagnosis. A step-wise, informed approach to post-transplant diarrhea will help the clinician achieve the best diagnostic yield. The use of diagnostic endoscopy should be preceded by exclusion of an infectious or drug-related cause of diarrhea. Empiric management with antidiarrheal agents, probiotics, and lactose-free diets may have a role in managing patients for whom no cause can be determined even after an extensive investigation. CONCLUSIONS Physicians should be familiar with the common etiologies that result in post-transplant diarrhea. A directed approach to diagnosis and treatment will not only help to resolve the diarrhea but also prevent potentially life-threatening consequences including loss of the graft as well. Prospective studies are required to determine the etiology of post-transplant diarrhea in different clinical and geographic settings.
Collapse
Affiliation(s)
- Chaitanya Pant
- University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| | | | | | | | | | | |
Collapse
|
34
|
MUNTEAN ADRIANA, LUCAN MIHAI. Immunosuppression in kidney transplantation. CLUJUL MEDICAL (1957) 2013; 86:177-80. [PMID: 26527942 PMCID: PMC4462507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 05/09/2013] [Indexed: 11/24/2022]
Abstract
For most patients with chronic kidney failure, kidney transplantation has the greatest potential for restoring a healthy and productive life. The risk of acute rejection is the highest in the first months after transplantation (induction phase) and diminishes afterwards (maintenance phase). Immunosuppression should be at the highest level in the early period and reduced for long-term therapy. At present, conventional immunosuppressive protocols consist of the triple therapy: a calcineurin inhibitor, an adjunctive agent, corticosteroids. The development of new immunosuppressives drugs is aimed not only at improving short-term outcomes, but also achieving better safety, less nephrotoxicity and minimal side effects.
Collapse
|
35
|
Ali AK. Pharmacovigilance Analysis of Serious Adverse Events Reported for Biologic Response Modifiers Used as Prophylaxis against Transplant Rejection: a Real-World Postmarketing Experience from the US FDA Adverse Event Reporting System (FAERS). Int J Organ Transplant Med 2013; 4:62-71. [PMID: 25013655 PMCID: PMC4089314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Immunosuppression by biologic response modifiers (BRM) is a crucial component for successful organ transplantation. In addition to their variable effectiveness in the prevention of organ rejection, these medications have safety concerns that complicate therapeutic outcomes in organ transplant patients. OBJECTIVE This study aims at identifying and characterizing safety signals of serious adverse events associated with exposure to BRM among organ transplant patients in a real-world environment. METHODS The FDA Adverse Event Reporting System was utilized to apply a pharmacovigilance disproportionality analysis to indentify serious adverse events. Associations between drugs and events were measured by empirical Bayes geometric mean (EBGM) and the corresponding 95% confidence intervals (EB05-EB95). Associations with EBGM≥2 were considered significant safety signals. RESULTS From 1997 to 2012, a total of 12,151 serious adverse event reports for BRM were reported; 15.6% of them (n=1,711) met the safety signal threshold of EB05>1, and 11.6% of these signals (n=199) were significant (EBGM≥2). Sirolimus and mycophenolate accounted for the majority of all signals; antithymocyte immunoglobulin (ATI) and cyclosporine contributed to the majority of significant signals. The following significant signals were identified for ATI (reduced therapeutic response, pulmonary edema, hypotension, serum sickness, infusion-related reaction, and anaphylactic reaction); for azathioprine (alternaria infection, fungal skin infection, and lymphoproliferative disorder); for cyclosporine (neurotoxicity, graft vs. host disease, and thyroid cancer); for cyclophosphamide (disease progression); for daclizumab (cytomegalovirus infection); and for tacrolimus (coma and tremor). 33.6% of these events contributed to patient death (n=67); 6.5% were life-threatening (n=13); 32.1% lead to hospitalization (n=64); and 27.6% resulted in other serious outcomes (n=55). CONCLUSION Utilization of BRM for the prophylaxis against transplant rejection is associated with serious adverse events that could be fatal.
Collapse
Affiliation(s)
- A. K. Ali
- Correspondence: Francis L. Delmonico, MD, Professor of Surgery, Harvard Medical School, Massachusetts General Hospital, Transplant Center, Boston, MA 02114–2696, USA , Tel: +1-617-726-2825 , E-mail:
| |
Collapse
|
36
|
Chang DJ, Lee S, Jang J, Kim SO, Kim WJ, Suh YG. Part II. Development of novel colchicine-derived immunosuppressants with improved pharmacokinetic properties. Bioorg Med Chem Lett 2012; 22:6750-5. [DOI: 10.1016/j.bmcl.2012.08.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 08/15/2012] [Accepted: 08/17/2012] [Indexed: 10/28/2022]
|
37
|
Zhou H, Lin J, Chen S, Ma L, Qiu Z, Chen W, Zhang X, Zhang Y, Lin X. Use of the ImmuKnow assay to evaluate the effect of alemtuzumab-depleting induction therapy on cell-mediated immune function after renal transplantation. Clin Exp Nephrol 2012; 17:304-9. [PMID: 23053591 DOI: 10.1007/s10157-012-0688-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 08/14/2012] [Indexed: 01/29/2023]
Abstract
BACKGROUND Good outcomes after renal transplantation are dependent on effective immunosuppression while minimizing infection. Alemtuzumab (Campath or Campath-1H) is an anti-CD52 humanized monoclonal IgG1 antibody which induces rapid and sustained depletion of circulating lymphocytes and has been effectively used as an immunosuppressant in post-transplant induction therapy. METHODS We used the ImmuKnow assay to compare cell-mediated immune function in renal transplant patients treated with alemtuzumab or with conventional immunosuppressive tri-therapy. The ImmuKnow method determines the levels of adenosine triphosphate (ATP) released from CD4 cells following stimulation with a mitogen. RESULTS We showed a statistically significant difference in the distribution of outcome after transplantation between the conventional and the Campath groups (P = 0.010). A significantly higher number of patients treated with alemtuzumab induction therapy were stable after transplantation compared to those treated with conventional immunosuppressive tri-therapy (96.6 vs. 75.7 %). ATP values were significantly higher in the conventional group compared to the Campath group at 180 days after transplantation (P < 0.001). ATP levels did not change significantly over time in clinically stable kidney recipients treated with alemtuzumab induction therapy (P = 0.554). CONCLUSIONS The ImmuKnow assay is a useful tool for evaluating the global immune response in alemtuzumab-treated renal transplant patients. Alemtuzumab-depleting induction therapy remains effective for at least 180 days.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Urology, People's Hospital Affiliated to Fujian University of Traditional Chinese Medicine (The People's Hospital of Fujian Province), No. 602, Middle Road 817, Fuzhou, 350004, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Therapeutic monoclonal antibodies (mAbs) are currently being approved for marketing in Europe and the United States, as well as other countries, on a regular basis. As more mAbs become available to physicians and patients, keeping track of the number, types, production cell lines, antigenic targets, and dates and locations of approvals has become challenging. Data are presented here for 34 mAbs that were approved in either Europe or the United States (US) as of March 2012, and nimotuzumab, which is marketed outside Europe and the US. Of the 34 mAbs, 28 (abciximab, rituximab, basiliximab, palivizumab, infliximab, trastuzumab, alemtuzumab, adalimumab, tositumomab-I131, cetuximab, ibrituximab tiuxetan, omalizumab, bevacizumab, natalizumab, ranibizumab, panitumumab, eculizumab, certolizumab pegol, golimumab, canakinumab, catumaxomab, ustekinumab, tocilizumab, ofatumumab, denosumab, belimumab, ipilimumab, brentuximab) are currently marketed in Europe or the US. Data for six therapeutic mAbs (muromonab-CD3, nebacumab, edrecolomab, daclizumab, gemtuzumab ozogamicin, efalizumab) that were approved but have been withdrawn or discontinued from marketing in Europe or the US are also included.
Collapse
|
39
|
Klipa D, Mahmud N, Ahsan N. Antibody immunosuppressive therapy in solid organ transplant: Part II. MAbs 2010; 2:607-12. [PMID: 20948291 DOI: 10.4161/mabs.2.6.13586] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The use of antibodies in transplantation dates to 1986 when muromonab CD3, a monoclonal antibody (mAb) targeting CD3, was first approved for prevention and treatment of renal allograft rejection. These agents have largely been used in a brief adjunctive manner to provide immunosuppression during the initial period after solid organ transplantation or during an episode of acute rejection. Recent advances in our understanding of transplant immunology have allowed emergence of numerous new mAbs, targeting co-stimulatory signals, cell surface receptors and novel protein constructs. During the next decade, transplant professionals will increasingly require knowledge of the mechanisms and pharmacologic characteristics of these novel therapeutic agents.
Collapse
|
40
|
Li J, Zhou B, Shentu J, Du L, Tan M, Hou S, Qian W, Li B, Zhang D, Dai J, Wang H, Zhang X, Chen J, Guo Y. Phase I trial of a humanized, Fc receptor nonbinding anti-CD3 antibody, hu12F6mu in patients receiving renal allografts. MAbs 2010; 2:449-56. [PMID: 20519962 DOI: 10.4161/mabs.12305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Hu12F6mu is an Fc-mutated, humanized anti-CD3 antibody developed in our lab. The aim of this study was to assess single dose escalation pharmacokinetics (PK) and safety profile of hu12F6mu and to measure the effects of the antibody on levels of circulating T cells over time. Twenty-seven patients receiving renal allografts were randomized to receive hu12F6mu intravenously at a single-dose of 2.5, 5 or 10 mg. The concentration-time data obtained by a validated ELISA method were subjected to non-compartmental PK analysis by DAS 2.1 software. Subgroups of CD2(+), CD3(+), CD4(+) and CD8(+) lymphocytes were monitored periodically by flow cytometry. Our results showed that hu12F6mu exhibited linear PK over the dose range of 2.5 to 10 mg. A significant decline in the proportion of T cells was observed immediately after the infusion, followed by a progressive increase occurring over the ensuing days of therapy. A significant negative correlation was observed between serum concentration of hu12F6mu and CD3(+) cell proportion. Intravenous infusion of hu12F6mu was well-tolerated in patients receiving renal allografts. These results suggest that hu12F6mu may have potential as a therapeutic agent, although further studies are needed.
Collapse
Affiliation(s)
- Jing Li
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | - Bo Zhou
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | | | - Li Du
- PLA Postgraduate School of Medicine; Second Military Medical University, China
| | - Min Tan
- PLA Postgraduate School of Medicine; Second Military Medical University, China
| | - Sheng Hou
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | - Weizhu Qian
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | - Bohua Li
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | - Dapeng Zhang
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | - Jianxin Dai
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | - Hao Wang
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| | - Xu Zhang
- PLA Postgraduate School of Medicine, China
| | | | - Yajun Guo
- PLA Postgraduate School of Medicine; Second Military Medical University; National Engineering Research Center of Antibody Medicine, China
| |
Collapse
|
41
|
Li J, Zhou B, Shentu J, Du L, Tan M, Hou S, Qian W, Li B, Zhang D, Dai J, Wang H, Zhang X, Chen J, Guo Y. Phase I trial of a humanized, Fc receptor nonbinding anti-CD3 antibody, hu12F6mu in patients receiving renal allografts. MAbs 2010; 2. [PMID: 20519962 PMCID: PMC3180091 DOI: 10.4161/mabs.2.4.12305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Hu12F6mu is an Fc-mutated, humanized anti-CD3 antibody developed in our lab. The aim of this study was to assess single dose escalation pharmacokinetics (PK) and safety profile of hu12F6mu and to measure the effects of the antibody on levels of circulating T cells over time. Twenty-seven patients receiving renal allografts were randomized to receive hu12F6mu intravenously at a single-dose of 2.5, 5 or 10 mg. The concentration-time data obtained by a validated ELISA method were subjected to non-compartmental PK analysis by DAS 2.1 software. Subgroups of CD2(+), CD3(+), CD4(+) and CD8(+) lymphocytes were monitored periodically by flow cytometry. Our results showed that hu12F6mu exhibited linear PK over the dose range of 2.5 to 10 mg. A significant decline in the proportion of T cells was observed immediately after the infusion, followed by a progressive increase occurring over the ensuing days of therapy. A significant negative correlation was observed between serum concentration of hu12F6mu and CD3(+) cell proportion. Intravenous infusion of hu12F6mu was well-tolerated in patients receiving renal allografts. These results suggest that hu12F6mu may have potential as a therapeutic agent, although further studies are needed.
Collapse
Affiliation(s)
- Jing Li
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Bo Zhou
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Jianzhong Shentu
- First Affiliated Hospital, College of Medicine; Zhejiang University; Hangzhou, China
| | - Li Du
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China
| | - Min Tan
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China
| | - Sheng Hou
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Weizhu Qian
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Bohua Li
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Dapeng Zhang
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Jianxin Dai
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Hao Wang
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| | - Xu Zhang
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China
| | - Jianghua Chen
- First Affiliated Hospital, College of Medicine; Zhejiang University; Hangzhou, China
| | - Yajun Guo
- PLA General Hospital Cancer Center and Department of Urology; The PLA Postgraduate School of Medicine; Beijing, China,International Cancer Research Institute; The Second Military Medical University; Shanghai, China,National Engineering Research Center of Antibody Medicine; Shanghai, China
| |
Collapse
|