1
|
Henrich TJ, Bosch RJ, Godfrey C, Mar H, Nair A, Keefer M, Fichtenbaum C, Moisi D, Clagett B, Buck AM, Deitchman AN, Aweeka F, Li JZ, Kuritzkes DR, Lederman MM, Hsue PY, Deeks SG. Sirolimus reduces T cell cycling, immune checkpoint marker expression, and HIV-1 DNA in people with HIV. Cell Rep Med 2024; 5:101745. [PMID: 39321793 PMCID: PMC11513808 DOI: 10.1016/j.xcrm.2024.101745] [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: 11/12/2023] [Revised: 04/10/2024] [Accepted: 08/28/2024] [Indexed: 09/27/2024]
Abstract
Key HIV cure strategies involve reversing immune dysfunction and limiting the proliferation of infected T cells. We evaluate the safety of sirolimus, a mammalian target of rapamycin (mTOR) inhibitor, in people with HIV (PWH) and study the impact of sirolimus on HIV-1 reservoir size and HIV-1-specific immunity in a single-arm study of 20 weeks of treatment in PWH on antiretroviral therapy (ART). Sirolimus treatment does not impact HIV-1-specific CD8 T cell responses but leads to a significant decrease in CD4+ T cell-associated HIV-1 DNA levels at 20 weeks of therapy in the primary efficacy population (n = 16; 31% decline, p = 0.008). This decline persists for at least 12 weeks following cessation of the study drug. Sirolimus treatment also leads to a significant reduction in CD4+ T cell cycling and PD-1 expression on CD8+ lymphocytes. These data suggest that homeostatic proliferation of infected cells, an important mechanism for HIV persistence, is an intriguing therapeutic target.
Collapse
Affiliation(s)
- Timothy J Henrich
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA.
| | - Ronald J Bosch
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Catherine Godfrey
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Hanna Mar
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Apsara Nair
- Frontier Science and Technology Research Foundation, Amherst, NY 14226, USA
| | - Michael Keefer
- Department of Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | - Carl Fichtenbaum
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Daniela Moisi
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian Clagett
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Amanda M Buck
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA; San Francisco State University, San Francisco, CA 94132, USA
| | - Amelia N Deitchman
- Department of Clinical Pharmacology, University of California San Francisco, San Francisco, CA 94110, USA
| | - Francesca Aweeka
- Department of Clinical Pharmacology, University of California San Francisco, San Francisco, CA 94110, USA
| | - Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel R Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael M Lederman
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Priscilla Y Hsue
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - Steven G Deeks
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
2
|
Xu W, Chen H, Xiao H. mTORC2: A neglected player in aging regulation. J Cell Physiol 2024:e31363. [PMID: 38982866 DOI: 10.1002/jcp.31363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/21/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024]
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that plays a pivotal role in various biological processes, through integrating external and internal signals, facilitating gene transcription and protein translation, as well as by regulating mitochondria and autophagy functions. mTOR kinase operates within two distinct protein complexes known as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which engage separate downstream signaling pathways impacting diverse cellular processes. Although mTORC1 has been extensively studied as a pro-proliferative factor and a pro-aging hub if activated aberrantly, mTORC2 received less attention, particularly regarding its implication in aging regulation. However, recent studies brought increasing evidence or clues for us, which implies the associations of mTORC2 with aging, as the genetic elimination of unique subunits of mTORC2, such as RICTOR, has been shown to alleviate aging progression in comparison to mTORC1 inhibition. In this review, we first summarized the basic characteristics of mTORC2, including its protein architecture and signaling network. We then focused on reviewing the molecular signaling regulation of mTORC2 in cellular senescence and organismal aging, and proposed the multifaceted regulatory characteristics under senescent and nonsenescent contexts. Next, we outlined the research progress of mTOR inhibitors in the field of antiaging and discussed future prospects and challenges. It is our pleasure if this review article could provide meaningful information for our readers and call forth more investigations working on this topic.
Collapse
Affiliation(s)
- Weitong Xu
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Honghan Chen
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- The Lab of Aging Research, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Ponticelli C, Reggiani F, Moroni G. Autophagy: A Silent Protagonist in Kidney Transplantation. Transplantation 2024; 108:1532-1541. [PMID: 37953477 DOI: 10.1097/tp.0000000000004862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Autophagy is a lysosome-dependent regulated mechanism that recycles unnecessary cytoplasmic components. It is now known that autophagy dysfunction may have a pathogenic role in several human diseases and conditions, including kidney transplantation. Both defective and excessive autophagy may induce or aggravate several complications of kidney transplantation, such as ischemia-reperfusion injury, alloimmune response, and immunosuppressive treatment and side effects. Although it is still complicated to measure autophagy levels in clinical practice, more attention should be paid to the factors that may influence autophagy. In kidney transplantation, the association of low doses of a mammalian target of rapamycin inhibitor with low doses of a calcineurin inhibitor may be of benefit for autophagy modulation. However, further studies are needed to explore the role of other autophagy regulators.
Collapse
Affiliation(s)
| | - Francesco Reggiani
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Gabriella Moroni
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
4
|
Mansur J, Chang-Dávila D, Simões MG, Cristelli MP, Stopa Martins SB, de Sousa Proença HM, Viana LA, Ferreira AN, Doher MP, Medina-Pestana J, Mastroianni Kirsztajn G, Tedesco-Silva H. Multiple-target Therapy for Posttransplant Focal Segmental Glomerulosclerosis. Transplant Direct 2024; 10:e1651. [PMID: 38817628 PMCID: PMC11139459 DOI: 10.1097/txd.0000000000001651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 03/13/2024] [Indexed: 06/01/2024] Open
Abstract
Background There is no consensus on the ideal strategy to treat posttransplant focal segmental glomerulosclerosis. The multiple-target therapy, which consisted of high-dose intravenous cyclosporine, prednisone, and plasmapheresis, showed favorable results. Methods This single-center, prospective study sought to evaluate the multiple-target therapy in an independent cohort of patients. Results Thirteen patients with posttransplant focal segmental glomerulosclerosis received multiple-target therapy. Complete remission was achieved in 2 patients (15.4%), and partial remission in another 2 patients (15.4%). Four patients (30.7%) did not show remission, and 5 patients (38%) lost the graft because of posttransplant focal segmental glomerulosclerosis during the 12-mo follow-up. Premature discontinuation of treatment occurred in 10 patients (77%), all associated with infectious adverse events. Cytomegalovirus was the most common complication, and preemptive therapy was used instead of prophylaxis. Conclusions In this cohort of patients, the efficacy of the multiple-target therapy was poor and limited by the high incidence of infectious adverse events.
Collapse
Affiliation(s)
- Juliana Mansur
- Nephrology Division, Hospital do Rim, Fundação Oswaldo Ramos, São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | - Marcela Giraldes Simões
- Nephrology Division, Hospital do Rim, Fundação Oswaldo Ramos, São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | | | | | - Laila Almeida Viana
- Nephrology Division, Hospital do Rim, Fundação Oswaldo Ramos, São Paulo, Brazil
| | | | | | - José Medina-Pestana
- Nephrology Division, Hospital do Rim, Fundação Oswaldo Ramos, São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | - Helio Tedesco-Silva
- Nephrology Division, Hospital do Rim, Fundação Oswaldo Ramos, São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| |
Collapse
|
5
|
Evans JB, Chou L, Kaeberlein M, Promislow DE, Creevy KE. Case report: Severe asymptomatic hypertriglyceridemia associated with long-term low-dose rapamycin administration in a healthy middle-aged Labrador retriever. Front Vet Sci 2023; 10:1285498. [PMID: 38094495 PMCID: PMC10716302 DOI: 10.3389/fvets.2023.1285498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/02/2023] [Indexed: 02/01/2024] Open
Abstract
Rapamycin is an mTOR inhibitor that has been shown to extend the lifespan of laboratory model organisms. In humans, rapamycin is used at higher doses as an immunosuppressive medication to prevent organ rejection. Numerous adverse effects are seen with rapamycin treatment in humans, with one of the most common being dysregulation of lipid metabolism. In humans, this often manifests as mild to moderate serum lipid elevations, with a small subset developing extreme triglyceride elevations. This case report describes an eight-year-old, castrated male, clinically healthy Labrador retriever who developed severe hypertriglyceridemia associated with low-dose rapamycin administration over a six-month period. During this time, the dog was asymptomatic and displayed no other clinical abnormalities, aside from a progressive lipemia. Within 15 days of discontinuing rapamycin treatment, and with no targeted lipemic intervention, the dog's lipemia and hypertriglyceridemia completely resolved.
Collapse
Affiliation(s)
- Jeremy B. Evans
- Department of Small Animal Clinical Sciences, Texas A&M School of Veterinary Medicine & Biomedical Sciences, College Station, TX, United States
| | - Lucy Chou
- Department of Small Animal Clinical Sciences, Texas A&M School of Veterinary Medicine & Biomedical Sciences, College Station, TX, United States
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States
- Optispan, Inc., Seattle, WA, United States
| | - Daniel E.L. Promislow
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, United States
- Department of Biology, University of Washington, Seattle, WA, United States
| | - Kate E. Creevy
- Department of Small Animal Clinical Sciences, Texas A&M School of Veterinary Medicine & Biomedical Sciences, College Station, TX, United States
| |
Collapse
|
6
|
Ponticelli C, Moroni G, Reggiani F. Autophagy and podocytopathy. Nephrol Dial Transplant 2023; 38:1931-1939. [PMID: 36708169 DOI: 10.1093/ndt/gfad024] [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: 11/19/2022] [Indexed: 01/29/2023] Open
Abstract
Autophagy is a complex process of lysosomal-dependent degradation of unwanted cellular material. In response to endogenous or exogenous stimuli, autophagy is induced and regulated by two kinases: the AMP activated kinase and the mammalian target of rapamycin (mTOR). Cells activated by Unc-51-like kinase 1 form a double membrane complex that sequesters the cargo (phagophore) and elongates producing spherical vesicles (autophagosomes). These reach and fuse with lysosomes, which degrade the cargo (autolysosomes). The resulting macromolecules are released back and recycled in the cytosol for reuse. In the podocyte, autophagy is a homeostatic mechanism that contributes to the formation and preservation of the morphological and functional integrity of actin cytoskeleton. Podocytes, fenestrated endothelial cells and glomerular basement membrane compose the glomerular filtration barrier. Podocyte damage may cause dysfunction of the glomerular barrier, proteinuria and glomerulosclerosis in different glomerular diseases and particularly in so-called podocytopathies, namely minimal change disease and focal segmental glomerulosclerosis. Several drugs and molecules may activate autophagic function in murine models. Among them, aldosterone inhibitors, mineralocorticoid inhibitors and vitamin D3 were proven to protect podocyte from injury and reduce proteinuria in clinical studies. However, no clinical trial with autophagy regulators in podocytopathies has been conducted. Caution is needed with other autophagy activators, such as mTOR inhibitors and metformin, because of potential adverse events.
Collapse
Affiliation(s)
| | - Gabriella Moroni
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Francesco Reggiani
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
7
|
Zhong Q, Xiao X, Qiu Y, Xu Z, Chen C, Chong B, Zhao X, Hai S, Li S, An Z, Dai L. Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm (Beijing) 2023; 4:e261. [PMID: 37143582 PMCID: PMC10152985 DOI: 10.1002/mco2.261] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Protein posttranslational modifications (PTMs) refer to the breaking or generation of covalent bonds on the backbones or amino acid side chains of proteins and expand the diversity of proteins, which provides the basis for the emergence of organismal complexity. To date, more than 650 types of protein modifications, such as the most well-known phosphorylation, ubiquitination, glycosylation, methylation, SUMOylation, short-chain and long-chain acylation modifications, redox modifications, and irreversible modifications, have been described, and the inventory is still increasing. By changing the protein conformation, localization, activity, stability, charges, and interactions with other biomolecules, PTMs ultimately alter the phenotypes and biological processes of cells. The homeostasis of protein modifications is important to human health. Abnormal PTMs may cause changes in protein properties and loss of protein functions, which are closely related to the occurrence and development of various diseases. In this review, we systematically introduce the characteristics, regulatory mechanisms, and functions of various PTMs in health and diseases. In addition, the therapeutic prospects in various diseases by targeting PTMs and associated regulatory enzymes are also summarized. This work will deepen the understanding of protein modifications in health and diseases and promote the discovery of diagnostic and prognostic markers and drug targets for diseases.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xina Xiao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yijie Qiu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhiqiang Xu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Chunyu Chen
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Baochen Chong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xinjun Zhao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shan Hai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shuangqing Li
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhenmei An
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Lunzhi Dai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
8
|
Galeev SR, Gautier SV. Risks and ways of preventing kidney dysfunction in drug-induced immunosuppression in solid organ recipients. RUSSIAN JOURNAL OF TRANSPLANTOLOGY AND ARTIFICIAL ORGANS 2022. [DOI: 10.15825/1995-1191-2022-4-24-38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunosuppressive therapy (IMT) is the cornerstone of treatment after transplantation. The goal of immunosuppression is to prevent acute and chronic rejection while maximizing patient survival and long-term graft function. However, the expected effects of IMT must be balanced against the major adverse effects of these drugs and their toxicity. The purpose of this review is to summarize world experience on current immunosuppressive strategies and to assess their effects on renal function.
Collapse
Affiliation(s)
- Sh. R. Galeev
- Shumakov National Medical Research Center of Transplantology and Artificial Organs
| | - S. V. Gautier
- Shumakov National Medical Research Center of Transplantology and Artificial Organs; Sechenov University
| |
Collapse
|
9
|
Ponticelli C, Citterio F. Non-Immunologic Causes of Late Death-Censored Kidney Graft Failure: A Personalized Approach. J Pers Med 2022; 12:1271. [PMID: 36013220 PMCID: PMC9410103 DOI: 10.3390/jpm12081271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 11/29/2022] Open
Abstract
Despite continuous advances in surgical and immunosuppressive protocols, the long-term survival of transplanted kidneys is still far from being satisfactory. Antibody-mediated rejection, recurrent autoimmune diseases, and death with functioning graft are the most frequent causes of late-kidney allograft failure. However, in addition to these complications, a number of other non-immunologic events may impair the function of transplanted kidneys and directly or indirectly lead to their failure. In this narrative review, we will list and discuss the most important nonimmune causes of late death-censored kidney graft failure, including quality of the donated kidney, adherence to prescriptions, drug toxicities, arterial hypertension, dyslipidemia, new onset diabetes mellitus, hyperuricemia, and lifestyle of the renal transplant recipient. For each of these risk factors, we will report the etiopathogenesis and the potential consequences on graft function, keeping in mind that in many cases, two or more risk factors may negatively interact together.
Collapse
Affiliation(s)
| | - Franco Citterio
- Renal Transplant Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
10
|
Relevance of NLRP3 Inflammasome-Related Pathways in the Pathology of Diabetic Wound Healing and Possible Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9687925. [PMID: 35814271 PMCID: PMC9262551 DOI: 10.1155/2022/9687925] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Wound healing is a major secondary complication in type 2 diabetes, which results in significant disability and mortality, imposing a significant clinical and social burden. Sustained activation of the Nod-like receptor protein (NLRP) inflammasome in wounds is responsible for excessive inflammatory responses and aggravates wound damage. The activation of the NLRP3 inflammasome is regulated by a two-step process: the priming/licensing (signal 1) step involved in transcription and posttranslation and the protein complex assembly (signal 2) step triggered by danger molecules. This review focuses on the advances made in understanding the pathophysiological mechanisms underlying wound healing in the diabetic microenvironment. Simultaneously, this review summarizes the molecular mechanisms of the main regulatory pathways associated with signal 1 and signal 2, which trigger the NLRP3 inflammasome complex assembly in the development of diabetic wounds (DW). Activation of the NLRP3 inflammasome-related pathway, involving the disturbance in Nrf2 and the NF-κB/NLRP3 inflammasome, TLR receptor-mediated activation of the NF-κB/NLRP3 inflammasome, and various stimuli inducing NLRP3 inflammasome assembly play a pivotal role in DW healing. Furthermore, therapeutics targeting the NLRP3 inflammasome-related pathways may promote angiogenesis, reprogram immune cells, and improve DW healing.
Collapse
|
11
|
Podestà MA, Faravelli I, Ponticelli C. Autophagy in lupus nephritis: A delicate balance between regulation and disease. Clin Exp Rheumatol 2022; 21:103132. [PMID: 35690243 DOI: 10.1016/j.autrev.2022.103132] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/07/2022] [Indexed: 11/02/2022]
Abstract
Autophagy is a highly regulated process wherein an unwanted cargo of damaged and dysfunctional cytoplasmic components is removed, delivered to lysosomes for degradation, and released back into the cytoplasm. Accumulating evidence suggests an important role of autophagy in the pathophysiology of systemic lupus erythematosus, with profound effects on both innate and adaptive immunity. Autophagy downregulation results in the inhibition of antigen presenting cells, reduced release of neutrophil extracellular traps and decreased activation of effector T and B cells, leading to reduced autoantibody production and attenuated type 1 interferon signaling. However, defective autophagy may accelerate the production of other inflammatory cytokines and reduce the clearance of apoptotic cells, promoting lupus development. In addition, autophagy dysfunction can concur to the pathogenesis of kidney injury in lupus nephritis. Autophagy is a pivotal mechanism to maintain podocyte integrity and endothelial cell survival. Several animal models have demonstrated that defective autophagy leads to podocyte injury and can promote an endothelial pro-inflammatory and atherogenic phenotype. Moreover, autophagy is a key homeostatic regulator of renal tubular cells, and recent evidence has pointed out that chronic autophagy deficiency may accelerate kidney fibrosis. Targeting autophagy may theoretically improve lupus nephritis outcomes, but novel, non-invasive methods to measure and monitor autophagic activity are urgently needed. In addition, the extent and timing of autophagy inhibition still require additional studies before clinical translation may be attempted. In this review, we will also discuss the effect of several clinically available drugs that can regulate the autophagic flux and their effect in lupus nephritis patients.
Collapse
Affiliation(s)
- Manuel Alfredo Podestà
- Renal Division, Department of Health Sciences, Università degli Studi di Milano, Milano, Italy.
| | - Irene Faravelli
- Neuroscience Section, Dino Ferrari Centre, Department of Pathophysiology and Transplantation, University of Milan, Milano, Italy
| | | |
Collapse
|
12
|
Gago LG, Jarrín DA, Magariños CR, Rodríguez MC, Hermida TF, Muñiz AL, Rivera CF, Hernández ÁA. Edema Associated With Everolimus de Novo. Transplant Proc 2021; 53:2681-2684. [PMID: 34620498 DOI: 10.1016/j.transproceed.2021.07.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/16/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND The appearance of edema limits the use of everolimus de novo together with tacrolimus and steroids in kidney transplantation. We aimed to investigate the frequency and characteristics of patients with edema and compare them according to the type of immunosuppression. METHODS We studied 150 kidney transplant recipients between 2015 and 2017 based on receiving everolimus de novo (group A) or mycophenolic acid derivatives (group B). RESULTS We analyzed 50 patients in group A and 100 in group B. Follow-up was 26.2 ± 10 months. Fifty-six patients presented edema (37.3%): 54% in group A and 29% in group B (P = .003). Edema was mild in 74% of patients in group A and 57.1% in group B. The probability of edema was 10.1%, 22.4%, and 41% at 3, 6, and 12 months, respectively, in group A vs 10.1%, 20.3%, and 25.4% in group B (P = .006). Patients were treated mostly with diuretics (14.3% in group A vs 27.6% in group B) and discontinuation of calcium channel blockers (46.4% in group A vs 48.3% in group B). Improvement was 70.4% in group A vs 60.7% in group B; patient worsening was 0% in group A vs 10.7% in group B; and there was no change in 29.6% in group A vs 28.6% in group B. We did not find differences in patient or graft survival in those who presented edema, regardless of the treatment group. CONCLUSION The use of everolimus and standard doses of tacrolimus caused edema in 54% of patients, with no impact on renal function or survival compared with mycophenolic acid derivatives. The edema was mostly of low intensity and improved in most patients.
Collapse
|
13
|
Ponticelli C, Zaina B, Moroni G. Planned Pregnancy in Kidney Transplantation. A Calculated Risk. J Pers Med 2021; 11:jpm11100956. [PMID: 34683097 PMCID: PMC8537874 DOI: 10.3390/jpm11100956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Pregnancy is not contraindicated in kidney transplant women but entails risks of maternal and fetal complications. Three main conditions can influence the outcome of pregnancy in transplant women: preconception counseling, maternal medical management, and correct use of drugs to prevent fetal toxicity. Preconception counseling is needed to prevent the risks of an unplanned untimely pregnancy. Pregnancy should be planned ≥2 years after transplantation. The candidate for pregnancy should have normal blood pressure, stable serum creatinine <1.5 mg/dL, and proteinuria <500 mg/24 h. Maternal medical management is critical for early detection and treatment of complications such as hypertension, preeclampsia, thrombotic microangiopathy, graft dysfunction, gestational diabetes, and infection. These adverse outcomes are strongly related to the degree of kidney dysfunction. A major issue is represented by the potential fetotoxicity of drugs. Moderate doses of glucocorticoids, azathioprine, and mTOR inhibitors are relatively safe. Calcineurin inhibitors (CNIs) are not associated with teratogenicity but may increase the risk of low birth weight. Rituximab and eculizumab should be used in pregnancy only if the benefits outweigh the risk for the fetus. Renin-angiotensin system inhibitors, mycophenolate, bortezomib, and cyclophosphamide can lead to fetal toxicity and should not be prescribed to pregnant women.
Collapse
Affiliation(s)
- Claudio Ponticelli
- Nephrology, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence:
| | - Barbara Zaina
- Department of Obstetrics and Gynecology, IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Gabriella Moroni
- Department of Biomedical Sciences, IRCCS Humanitas Research Hospital, Humanitas University, 20122 Milan, Italy;
| |
Collapse
|
14
|
Hoff U, Markmann D, Nieminen-Kelhä M, Budde K, Hegner B. Low-dose rapamycin does not impair vascular integrity and tubular regeneration after kidney transplantation in rats. Sci Rep 2021; 11:16270. [PMID: 34381142 PMCID: PMC8358014 DOI: 10.1038/s41598-021-95790-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/16/2021] [Indexed: 11/23/2022] Open
Abstract
mTOR inhibitors offer advantages after kidney transplantation including antiviral and antitumor activity besides facilitating low calcineurin inhibitor exposure to reduce nephrotoxicity. Concerns about adverse effects due to antiproliferative and antiangiogenic properties have limited their clinical use particularly early after transplantation. Interference with vascular endothelial growth factor (VEGF)-A, important for physiologic functioning of renal endothelial cells and tubular epithelium, has been implicated in detrimental renal effects of mTOR inhibitors. Low doses of Rapamycin (loading dose 3 mg/kg bodyweight, daily doses 1.5 mg/kg bodyweight) were administered in an allogenic rat kidney transplantation model resulting in a mean through concentration of 4.30 ng/mL. Glomerular and peritubular capillaries, tubular cell proliferation, or functional recovery from preservation/reperfusion injury were not compromised in comparison to vehicle treated animals. VEGF-A, VEGF receptor 2, and the co-receptor Neuropilin-1 were upregulated by Rapamycin within 7 days. Rat proximal tubular cells (RPTC) responded in vitro to hypoxia with increased VEGF-A and VEGF-R1 expression that was not suppressed by Rapamycin at therapeutic concentrations. Rapamycin did not impair proliferation of RPTC under hypoxic conditions. Low-dose Rapamycin early posttransplant does not negatively influence the VEGF network crucial for recovery from preservation/reperfusion injury. Enhancement of VEGF signaling peritransplant holds potential to further improve outcomes.
Collapse
Affiliation(s)
- Uwe Hoff
- Department of Nephrology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Denise Markmann
- Nieren- und Dialysezentrum Schöneberg-Tempelhof, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Björn Hegner
- Department of Nephrology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Vitanas Hospital for Geriatric Medicine, Berlin, Germany.
| |
Collapse
|
15
|
Henrich TJ, Schreiner C, Cameron C, Hogan LE, Richardson B, Rutishauser RL, Deitchman AN, Chu S, Rogers R, Thanh C, Gibson EA, Zarinsefat A, Bakkour S, Aweeka F, Busch MP, Liegler T, Baker C, Milush J, Deeks SG, Stock PG. Everolimus, an mTORC1/2 inhibitor, in ART-suppressed individuals who received solid organ transplantation: A prospective study. Am J Transplant 2021; 21:1765-1779. [PMID: 32780519 PMCID: PMC9177122 DOI: 10.1111/ajt.16244] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/30/2020] [Accepted: 07/19/2020] [Indexed: 01/25/2023]
Abstract
Pharmacologic inhibition of the mammalian target of rapamycin (mTOR) in the setting of renal transplantation has previously been associated with lower human immunodeficiency virus 1 (HIV-1) DNA burden, and in vitro studies suggest that mTOR inhibition may lead to HIV transcriptional silencing. Because prospective clinical trials are lacking, we conducted an open-label, single-arm study to determine the impact of the broad mTOR inhibitor, everolimus, on residual HIV burden, transcriptional gene expression profiles, and immune responses in HIV-infected adult solid organ transplant (SOT) recipients on antiretroviral therapy. Whereas everolimus therapy did not have an overall effect on cell-associated HIV-1 DNA and RNA levels in the entire cohort, participants who maintained everolimus time-averaged trough levels >5 ng/mL during the first 2 months of therapy had significantly lower RNA levels up to 6 months after the cessation of study drug. Time-averaged everolimus trough levels significantly correlated with greater inhibition of mTOR gene pathway transcriptional activity. Everolimus treatment also led to decreased PD-1 expression on certain T cell subsets. These data support the rationale for further study of the effects of mTOR inhibition on HIV transcriptional silencing in non-SOT populations, either alone or in combination with other strategies. Trial Registration: ClinicalTrials.gov NCT02429869.
Collapse
Affiliation(s)
- Timothy J. Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Corinna Schreiner
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA,Institute of Biochemistry and Molecular Biology, Ulm University, Germany
| | - Cheryl Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Louise E. Hogan
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Brian Richardson
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Rachel L. Rutishauser
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Amelia N. Deitchman
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA
| | - Simon Chu
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Rodney Rogers
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Cassandra Thanh
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Erica A. Gibson
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Arya Zarinsefat
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Francesca Aweeka
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA
| | | | - Teri Liegler
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Christopher Baker
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Jeffrey Milush
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases & Global Medicine, University of California San Francisco, San Francisco, CA
| | - Peter G. Stock
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
16
|
Weiss D, Teichler A, Hoeger PH. Long-term sirolimus treatment in blue rubber bleb nevus syndrome: Case report and review of the literature. Pediatr Dermatol 2021; 38:464-468. [PMID: 33382463 DOI: 10.1111/pde.14506] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Blue rubber bleb nevus syndrome is a rare vascular syndrome characterized by continuous eruption of vascular nodules in the skin, mucous membranes, and solid organs due to somatic activating mutations of the angiopoietin receptor TEK gene. It may be complicated by acute life-threatening hemorrhage and localized intravascular coagulation. We report an 11-year-old girl with complicated blue rubber bleb nevus syndrome treated with sirolimus since the age of 2. We review the literature on sirolimus therapy for children with blue rubber bleb nevus syndrome.
Collapse
Affiliation(s)
- Doris Weiss
- Department of Paediatric Dermatology, Catholic Children's Hospital Wilhelmstift, Hamburg, Germany.,Department of Dermatology, Medical University of Vienna, Wien, Austria
| | - Anne Teichler
- Department of Paediatric Dermatology, Catholic Children's Hospital Wilhelmstift, Hamburg, Germany
| | - Peter H Hoeger
- Department of Paediatric Dermatology, Catholic Children's Hospital Wilhelmstift, Hamburg, Germany.,Department of Paediatrics, Catholic Children's Hospital Wilhelmstift, Hamburg, Germany
| |
Collapse
|
17
|
Long-term follow-up of mTOR inhibition for Erdheim-Chester disease. Blood 2021; 135:1994-1997. [PMID: 32299103 DOI: 10.1182/blood.2019004478] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Two articles this week focus on Erdheim-Chester disease (ECD), a rare histiocytosis that mainly affects adults. Clonal somatic mutations primarily involving proteins in the BRAF and MPAK pathways have established ECD as a myeloid neoplasm, with targeted therapies now available for patients. In the first paper, an international panel presents new consensus recommendations for evaluation and treatment of ECD. In the second paper, Pegoraro and colleagues present long-term outcomes of patients with ECD treated with sirolimus, with responses in patients both with and without BRAF mutations.
Collapse
|
18
|
Ponticelli C, Arnaboldi L, Moroni G, Corsini A. Treatment of dyslipidemia in kidney transplantation. Expert Opin Drug Saf 2020; 19:257-267. [DOI: 10.1080/14740338.2020.1732921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Claudio Ponticelli
- Divisione di Nefrologia, Istituto Scientifico Ospedale Maggiore, Milano, Italy (retired)
| | - Lorenzo Arnaboldi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DISFeB), Università degli Studi di Milano, Milano, Italy
| | - Gabriella Moroni
- Nefrologia e Dialisi, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Alberto Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DISFeB), Università degli Studi di Milano, Milano, Italy
- IRCCS Multimedica, Milano, Italy
| |
Collapse
|
19
|
Low-Dose Sirolimus Immunoregulation Therapy in Patients with Active Rheumatoid Arthritis: A 24-Week Follow-Up of the Randomized, Open-Label, Parallel-Controlled Trial. J Immunol Res 2019; 2019:7684352. [PMID: 31781682 PMCID: PMC6874993 DOI: 10.1155/2019/7684352] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 08/30/2019] [Indexed: 01/01/2023] Open
Abstract
Background We have reported previously the insufficient absolute number or functional defects of regulatory T cells (Tregs) in patients with rheumatoid arthritis (RA), challenging conventional unspecific immunosuppressive therapy. Sirolimus, a mTOR inhibitor, is reported to allow growth of functional Tregs; here, we investigated the efficacy of low-dose sirolimus combined with conventional immunosuppressants (sirolimus immunoregulation therapy) for RA treatment with lower side effects and better tolerance. Methods In this nonblinded and parallel-group trial, we randomly assigned 62 patients to receive conventional glucocorticoids and immunosuppressants with or without sirolimus at a dosage of 0.5 mg on alternate days for 24 weeks in a 2 : 1 ratio. The demographic features, clinical manifestations, and laboratory indicators including peripheral blood lymphocyte subgroups and CD4+T subsets were compared before and after the treatment. Results Finally, 37 patients in the sirolimus group and 18 in the conventional treated group completed the 6-month study. By 24 weeks, the patients with sirolimus experienced significant reduction in disease activity indicators including DAS28, ESR, and the number of tender joints and swollen joints (p < 0.001). Notably, they had a higher level of Tregs as compared with those with conventional therapy alone (p < 0.05), indicating that sirolimus could partly restore the reduced Tregs. Concomitantly, their usage of immunosuppressants for controlling disease activity was decreased as compared with the conventional group with no difference in blood routine, and liver and renal functions both before and after the treatment of sirolimus and between the two groups (p > 0.05). Conclusions Low-dose sirolimus immunoregulatory therapy selectively upregulated Tregs and partly replaced the usage of immunosuppressants to control disease activity without overtreatment and evaluable side effect. Further study is required using a large sample of RA patients treated with sirolimus for a longer period. This trial is registered at the Chinese Clinical Trial Registry (http://www.chictr.org.cn/showproj.aspx?proj=17245).
Collapse
|
20
|
Ponticelli C, Moroni G. Fetal Toxicity of Immunosuppressive Drugs in Pregnancy. J Clin Med 2018; 7:jcm7120552. [PMID: 30558290 PMCID: PMC6306731 DOI: 10.3390/jcm7120552] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 01/20/2023] Open
Abstract
Women affected by autoimmune diseases, organ transplantation, or neoplasia need to continue immunosuppressive treatment during pregnancy. In this setting, not only a careful planning of pregnancy, but also the choice of drugs is critical to preventing maternal complications and minimizing the fetal risks. Some immunosuppressive drugs are teratogenic and should be replaced even before the pregnancy, while other drugs need to be managed with caution to prevent fetal risks, including miscarriage, intrauterine growth restriction, prematurity, and low birth weight. In particular, the increasing use of biologic agents raises the question of their compatibility with reproduction. In this review we present data on the indication and safety in pregnancy of the most frequently used immunosuppressive drugs.
Collapse
Affiliation(s)
- Claudio Ponticelli
- Former Director Renal Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Gabriella Moroni
- Nephrological Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| |
Collapse
|
21
|
Sitruk L, Couchoud C, Hourmant M, Tuppin P, Macher MA, Legeai C. [Description of immunosuppressive maintenance treatments post kidney transplant through the National System of Health Insurance]. Nephrol Ther 2018; 14:523-530. [PMID: 29887268 DOI: 10.1016/j.nephro.2018.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/02/2018] [Accepted: 03/18/2018] [Indexed: 01/08/2023]
Abstract
The aim of this study was to describe the drug dispensing of maintenance immunosuppression treatment in 2014 for patients who received a kidney transplant in 2012, based on the data of the French national health insurance and to compare those results with the information collected in the national database for kidney recipients. For each patient, are considered all drugs dispensing with their dates of issue, the molecules and their presentations (number of pills and dosage). Among 2463 transplanted adults in 2012, 73% have received tacrolimus monohydrate, 59% mycophenolate mofetil, 54% prednisone and 20% cyclosporin in 2014. The daily doses but not the number of tablets per day declined with age. The most frequent association was tacrolimus monohydrate-mycophenolate mofetil-steroids in 34% of the cases. The use of mTOR inhibitors, rare generally (7%), is more common in patients aged 66-85 years. Associations did not differ significantly according to diabetic status, for patients with a kidney from an elderly donor or according to the number of mismatch. The daily doses estimated from the deliverance in pharmacy are respectively similar, understated and over-estimate for tacrolimus, mycophenolate mofetil and cyclosporin compared to the national database. This study confirms the difficulty of apprehending drug consumption based only on dispensing in pharmacies or punctual recording even if it allows a fairly comprehensive view of French practices.
Collapse
Affiliation(s)
- Lola Sitruk
- Agence de la biomédecine, 1, avenue du Stade-de-France, 93212 Saint-Denis-La-Plaine cedex, France
| | - Cécile Couchoud
- Agence de la biomédecine, 1, avenue du Stade-de-France, 93212 Saint-Denis-La-Plaine cedex, France
| | - Maryvonne Hourmant
- Service de néphrologie et d'immunologie clinique, CHU, 30, boulevard Jean-Monnet, immeuble Jean-Monnet, 44093 Nantes cedex 1, France
| | - Philippe Tuppin
- Caisse nationale de l'Assurance maladie des travailleurs salariés, Direction de la stratégie des études et des statistiques, 26-50, avenue du Professeur-André-Lemierre, 75986 Paris cedex 20, France
| | - Marie-Alice Macher
- Agence de la biomédecine, 1, avenue du Stade-de-France, 93212 Saint-Denis-La-Plaine cedex, France
| | - Camille Legeai
- Agence de la biomédecine, 1, avenue du Stade-de-France, 93212 Saint-Denis-La-Plaine cedex, France.
| |
Collapse
|
22
|
Lai ZW, Kelly R, Winans T, Marchena I, Shadakshari A, Yu J, Dawood M, Garcia R, Tily H, Francis L, Faraone SV, Phillips PE, Perl A. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: a single-arm, open-label, phase 1/2 trial. Lancet 2018; 391:1186-1196. [PMID: 29551338 PMCID: PMC5891154 DOI: 10.1016/s0140-6736(18)30485-9] [Citation(s) in RCA: 282] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/22/2017] [Accepted: 11/28/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Patients with systemic lupus erythematosus have T-cell dysfunction that has been attributed to the activation of the mammalian target of rapamycin (mTOR). Rapamycin inhibits antigen-induced T-cell proliferation and has been developed as a medication under the generic designation of sirolimus. We assessed safety, tolerance, and efficacy of sirolimus in a prospective, biomarker-driven, open-label clinical trial. METHODS We did a single-arm, open-label, phase 1/2 trial of sirolimus in patients with active systemic lupus erythematosus disease unresponsive to, or intolerant of, conventional medications at the State University of New York Upstate Medical University (Syracuse, NY, USA). Eligible participants (aged ≥18 years) had active systemic lupus erythematosus fulfilling four or more of 11 diagnostic criteria defined by the American College of Rheumatology. We excluded patients with allergy or intolerance to sirolimus, patients with life-threatening manifestations of systemic lupus erythematosus, proteinuria, a urine protein to creatinine ratio higher than 0·5, anaemia, leucopenia, or thrombocytopenia. Patients received oral sirolimus at a starting dose of 2 mg per day, with dose adjusted according to tolerance and to maintain a therapeutic range of 6-15 ng/mL. Patients were treated with sirolimus for 12 months. Safety outcomes included tolerance as assessed by the occurrence of common side-effects. The primary efficacy endpoint was decrease in disease activity, assessed using the British Isles Lupus Assessment Group (BILAG) index and the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI). Blood samples of 56 matched healthy individuals were obtained as controls for immunobiological outcomes monitored at each visit. The primary efficacy endpoint was assessed in all patients who completed 12 months of treatment, and all patients who received at least one dose of treatment were included in the safety analyses. This trial is registered with ClinicalTrials.gov, number NCT00779194. FINDINGS Between March 9, 2009, and Dec 8, 2014, 43 patients were enrolled, three of whom did not meet eligibility criteria. 11 of the 40 eligible patients discontinued study treatment because of intolerance (n=2) or non-compliance (n=9). SLEDAI and BILAG disease activity scores were reduced during 12 months of treatment in 16 (55%) of 29 patients who completed treatment. Mean SLEDAI score decreased from 10·2 (SD 5·6) at enrolment to 4·8 (4·5) after 12 months of treatment (p<0·001) and the mean total BILAG index score decreased from 28·4 (12·4) at enrolment to 17·4 (10·7) after 12 months of treatment (p<0·001). The mean daily dose of prednisone required to control disease activity decreased from 23·7 mg (SD 9·6) to 7·2 mg (2·3; p<0·001) after 12 months of treatment. Sirolimus expanded CD4+CD25+FoxP3+ regulatory T cells and CD8+ memory T-cell populations and inhibited interleukin-4 and interleukin-17 production by CD4+ and CD4-CD8- double-negative T cells after 12 months. CD8+ memory T cells were selectively expanded in SRI-responders. Patient liver function and lymphocyte counts were unchanged. Although HDL-cholesterol (Z=-2·50, p=0·012), neutrophil counts (Z=-1·92, p=0·054), and haemoglobin (Z=-2·83, p=0·005) were moderately reduced during treatment, all changes occurred within a range that was considered safe. Platelet counts were slightly elevated during treatment (Z=2·06, p=0·0400). INTERPRETATION These data show that a progressive improvement in disease activity is associated with correction of pro-inflammatory T-cell lineage specification in patients with active systemic lupus erythematosus during 12 months of sirolimus treatment. Follow-up placebo-controlled clinical trials in diverse patient populations are warranted to further define the role of mTOR blockade in treatment of systemic lupus erythematosus. FUNDING Pfizer, the National Institutes of Health, and the Central New York Community Foundation.
Collapse
Affiliation(s)
- Zhi-Wei Lai
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ryan Kelly
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Thomas Winans
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ivan Marchena
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ashwini Shadakshari
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Julie Yu
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Maha Dawood
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ricardo Garcia
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Hajra Tily
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Lisa Francis
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Stephen V Faraone
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Paul E Phillips
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Andras Perl
- Division of Rheumatology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
23
|
Filler G, Licht C, Huang SHS. Is there a case for eculizumab for pediatric renal transplantation? Pediatr Transplant 2018; 22. [PMID: 29417722 DOI: 10.1111/petr.13128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Guido Filler
- Departments of Paediatrics and Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,The Lilibeth Caberto Kidney Clinical Research Unit, Western University, London, ON, Canada.,Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Christoph Licht
- Division of Nephrology and Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shih-Han Susan Huang
- Departments of Paediatrics and Medicine, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,The Lilibeth Caberto Kidney Clinical Research Unit, Western University, London, ON, Canada.,Department of Biophysics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
24
|
Lin Y, Wang B, Shan W, Tan Y, Feng J, Xu L, Wang L, Han B, Zhang M, Yu J, Yu X, Huang H. mTOR inhibitor rapamycin induce polymorphonuclear myeloid-derived suppressor cells mobilization and function in protecting against acute graft-versus-host disease after bone marrow transplantation. Clin Immunol 2017; 187:122-131. [PMID: 29132870 DOI: 10.1016/j.clim.2017.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 10/23/2017] [Accepted: 11/09/2017] [Indexed: 01/07/2023]
Abstract
The mammalian target of rapamycin (mTOR) inhibitor rapamycin (RAPA) has been shown to be an effective immunosuppressor in the management of acute graft-versus-host disease (aGVHD) after bone marrow transplantation. Myeloid-derived suppressor cells (MDSCs) also have a protective effect in aGVHD regulation. However, the relationship between RAPA and MDSCs in aGVHD models is unclear. Meanwhile, the effect of RAPA on different subgroups of MDSCs is also less well described. In this study, we demonstrate that in vivo administration of RAPA results in the expansion and functional enhancement of polymorphonuclear MDSCs (PMN-MDSCs) in a murine model of aGVHD. RAPA treatment can enhance the suppressive function of PMN-MDSCs via up-regulation of arginase1 (Arg1) and induced nitric oxide synthase (iNOS) at later time points. Moreover, RAPA can also induce a strong immunosuppressive function in PMN-MDSCs from murine bone marrow in vitro, but has a contrary effect on monocytic MDSCs (M-MDSCs). We found that RAPA-treated PMN-MDSCs can restrain the differentiation of Th1/Th2 cells and promote induction of regulatory T cells in in vitro studies.
Collapse
Affiliation(s)
- Yu Lin
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Binsheng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Shan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yamin Tan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Feng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Xu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Limengmeng Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China
| | - Biqing Han
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mingming Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Yu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohong Yu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
25
|
Filler G, Medeiros M. Improving long-term outcomes after pediatric renal transplantation by addressing dyslipidemia. Pediatr Transplant 2017; 21. [PMID: 28370889 DOI: 10.1111/petr.12880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Guido Filler
- Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Mara Medeiros
- Laboratorio de Investigacion en Nefrologia, Hospital Infantil de Mexico Federico Gomez, Mexico City, México.,Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, México
| |
Collapse
|
26
|
Uchida J, Iwai T, Kuwabara N, Kabei K, Nishide S, Yamasaki T, Naganuma T, Kumada N, Takemoto Y, Nakatanti T. Clinical Experience of Late Conversion From Antimetabolites With Standard Exposure Calcineurin Inhibitors to Everolimus With Calcineurin Inhibitor Minimization in Stable Kidney Transplant Recipients With Good Renal Function. Transplant Proc 2017; 48:775-80. [PMID: 27234734 DOI: 10.1016/j.transproceed.2016.02.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/30/2016] [Accepted: 02/18/2016] [Indexed: 12/13/2022]
Abstract
INTRODUCTION This study describes our clinical experience of late conversion from antimetabolites with standard exposure calcineurin inhibitors (CNIs) to everolimus with CNI minimization in stable kidney transplant recipients with good graft function. PATIENTS AND METHODS A 1-year retrospective pilot study of 26 kidney recipients converted from antimetabolites with standard exposure CNIs to everolimus with CNI minimization was performed. The recipients enrolled in this study had normal or slightly impaired renal function defined as a serum creatinine value <2.0 mg/dL, and normal or slightly increased albuminuria defined as a urinary albumin excretion rate <100 mg/g creatinine. RESULTS The median time from transplant to conversion was 39.5 months posttransplant (range, 3-275). Treatment with everolimus was stopped owing to adverse events in 11 patients (42.3%). In the analysis of the patients in whom everolimus was maintained, the mean estimated glomerular filtration rate (eGFR) significantly increased from 50.7 ± 11.9 mL/min/1.73 m(2) at baseline to 53.6 ± 13.9 mL/min/1.73 m(2) at 1 year after conversion. In the patients in whom everolimus was stopped during the observation period, there was no difference in eGFR between baseline and 1 year after conversion. CONCLUSIONS This study demonstrated that, among the patients converted to everolimus at a late stage, there was no deterioration in renal function whether everolimus was maintained or stopped within 1 year after conversion.
Collapse
Affiliation(s)
- J Uchida
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan.
| | - T Iwai
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - N Kuwabara
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - K Kabei
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - S Nishide
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - T Yamasaki
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - T Naganuma
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - N Kumada
- Department of Urology, Suita Municipal Hospital, Suita, Japan
| | - Y Takemoto
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - T Nakatanti
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
27
|
Lim LM, Kung LF, Kuo MC, Kuo HT. Risk Factors Associated With Mammalian Target of Rapamycin Inhibitor Withdrawal in Kidney Transplant Recipients. Transplant Proc 2016; 48:790-3. [PMID: 27234737 DOI: 10.1016/j.transproceed.2015.12.085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 12/30/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mammalian target of rapamycin inhibitors (mTORi) play an essential role as novel immunosuppressive agents in kidney transplantation (KT). Treatment cessation usually occurs after adverse effects occur. We investigated the risk factors associated with withdrawal of mTORi in KT recipients and evaluated the outcomes related to the withdrawal. METHODS The study enrolled KT recipients being followed up in a medical center in southern Taiwan from January 1999 through December 2014. RESULTS Risk factors associated with mTORi withdrawal were initial proteinuria level, higher initial serum creatinine level posttransplantation, and history of glomerulonephritis as the primary etiology of renal failure. mTORi withdrawal was associated with increased risk of graft failure (hazard ratio [HR], 9.97 [95% confidence interval (CI), 1.03-96.8]; P = .047). Higher body mass index (HR, 11.2 [95% CI, 1.63-76.6]; P = .01) and tacrolimus usage (HR, 8.30 [95% CI, 1.14-60.7]; P = .037) were associated with increased risk of new-onset diabetes after transplantation in mTORi withdrawal groups. CONCLUSIONS Proteinuria, poor graft function, and primary glomerulonephritis were associated with cessation of mTORi treatment. Earlier identification of these risk factors may prevent further adverse events and optimize transplantation outcomes after mTORi conversion.
Collapse
Affiliation(s)
- L M Lim
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - L F Kung
- Department of Nursing, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - M C Kuo
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Faculty of Renal Care, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - H T Kuo
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Faculty of Renal Care, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
28
|
Waldner M, Fantus D, Solari M, Thomson AW. New perspectives on mTOR inhibitors (rapamycin, rapalogs and TORKinibs) in transplantation. Br J Clin Pharmacol 2016; 82:1158-1170. [PMID: 26810941 DOI: 10.1111/bcp.12893] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 02/06/2023] Open
Abstract
The macrolide rapamycin and its analogues (rapalogs) constitute the first generation of mammalian target of rapamycin (mTOR) inhibitors. Since the introduction of rapamycin as an immunosuppressant, there has been extensive progress in understanding its complex mechanisms of action. New insights into the function of mTOR in different immune cell types, vascular endothelial cells and neoplastic cells have opened new opportunities and challenges regarding mTOR as a pharmacological target. Currently, the two known mTOR complexes, mTOR complex (mTORC) 1 and mTORC2, are the subject of intense investigation, and the introduction of second-generation dual mTORC kinase inhibitors (TORKinibs) and gene knockout mice is helping to uncover the distinct roles of these complexes in different cell types. While the pharmacological profiling of rapalogs is advanced, much less is known about the properties of TORKinibs. A potential benefit of mTOR inhibition in transplantation is improved protection against transplant-associated viral infections compared with standard calcineurin inhibitor-based immunosuppression. Preclinical and clinical data also underscore the potentially favourable antitumour effects of mTOR inhibitors in regard to transplant-associated malignancies and as a novel treatment option for various other cancers. Many aspects of the mechanisms of action of mTOR inhibitors and their clinical implications remain unknown. In this brief review we discuss new findings and perspectives of mTOR inhibitors in transplantation.
Collapse
Affiliation(s)
- Matthias Waldner
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Plastic Surgery, University of Zurich, Zurich, Switzerland
| | - Daniel Fantus
- Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mario Solari
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Angus W Thomson
- Starzl Transplantation Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Ventura-Aguiar P, Campistol JM, Diekmann F. Safety of mTOR inhibitors in adult solid organ transplantation. Expert Opin Drug Saf 2016; 15:303-19. [PMID: 26667069 DOI: 10.1517/14740338.2016.1132698] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Mammalian target of rapamycin (mTOR) inhibitors (sirolimus and everolimus) are a class of immunosuppressive drugs approved for solid organ transplantation (SOT). By inhibiting the ubiquitous mTOR pathway, they present a peculiar safety profile. The increased incidence of serious adverse events in early studies halted the enthusiasm as a kidney sparing alternative to calcineurin inhibitors (CNI). AREAS COVERED Herein we review mTOR inhibitors safety profile for adult organ transplantation, ranging from acute side effects, such as lymphoceles, delayed wound healing, or cytopenias, to long-term ones which increase morbidity and mortality, such as cancer risk and metabolic profile. Infection, proteinuria, and cutaneous safety profiles are also addressed. EXPERT OPINION In the authors' opinion, mTOR inhibitors are a safe alternative to standard immunosuppression therapy with CNI and mycophenolate/azathioprine. Mild adverse events can be easily managed with an increased awareness and close monitoring of trough levels. Most serious side effects are dose- and organ-dependent. In kidney and heart transplantation mTOR inhibitors may be safely used as either low-dose de novo or through early-conversion. In the liver, conversion 4 weeks post-transplantation may reduce long-term chronic kidney disease secondary to calcineurin nephrotoxicity, without increasing hepatic artery/portal vein thrombosis.
Collapse
Affiliation(s)
- Pedro Ventura-Aguiar
- a Department of Nephrology and Renal Transplantation , Hospital Clínic , Villarroel, 170, E-08036 Barcelona , Spain
| | - Josep Maria Campistol
- a Department of Nephrology and Renal Transplantation , Hospital Clínic , Villarroel, 170, E-08036 Barcelona , Spain.,b August Pi i Sunyer Biomedical Research Institute (IDIBAPS) , University of Barcelona , Barcelona , Spain
| | - Fritz Diekmann
- a Department of Nephrology and Renal Transplantation , Hospital Clínic , Villarroel, 170, E-08036 Barcelona , Spain
| |
Collapse
|
30
|
Pathophysiologic and treatment strategies for cardiovascular disease in end-stage renal disease and kidney transplantations. Cardiol Rev 2016; 23:109-18. [PMID: 25420053 DOI: 10.1097/crd.0000000000000044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The inextricable link between the heart and the kidneys predestines that significant cardiovascular disease ensues in the face of end-stage renal disease (ESRD). As a point of fact, the leading cause of mortality of patients on dialysis is still from cardiovascular etiologies, albeit differing in particular types of disease from the general population. For example, sudden cardiac death outnumbers coronary artery disease in patients with ESRD, which is the reverse for the general population. In this review, we will focus on the pathophysiology and treatment options of important traditional and nontraditional risk factors for cardiovascular disease in ESRD patients such as hypertension, anemia, vascular calcification, hyperparathyroidism, uremia, and oxidative stress. The evidence of erythropoietin-stimulating agents, phosphate binders, calcimimetics, and dialysis modalities will be presented. We will then discuss how these risk factors may be changed and perhaps exacerbated after renal transplantation. This is largely due to the immunosuppressive agents that are both crucial yet potentially detrimental in the posttransplant state. Calcineurin inhibitors, corticosteroids, and mammalian target of rapamycin inhibitors, the mainstay of transplant immunosuppression, are all known to increase the risks of developing new onset diabetes as well as the metabolic syndrome. Thus, we need to carefully negotiate between patients' cardiovascular profile and their risks of rejection. Finally, we end by considering strategies by which we may minimize cardiovascular disease in the transplant population, as this modality still confers the highest chance of survival in patients with ESRD.
Collapse
|
31
|
Rogers J, Katari R, Gifford S, Tamburrini R, Edgar L, Voigt MR, Murphy SV, Igel D, Mancone S, Callese T, Colucci N, Mirzazadeh M, Peloso A, Zambon JP, Farney AC, Stratta RJ, Orlando G. Kidney transplantation, bioengineering and regeneration: an originally immunology-based discipline destined to transition towards ad hoc organ manufacturing and repair. Expert Rev Clin Immunol 2015; 12:169-82. [PMID: 26634874 DOI: 10.1586/1744666x.2016.1112268] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Kidney transplantation (KT), as a modality of renal replacement therapy (RRT), has been shown to be both economically and functionally superior to dialysis for the treatment of end-stage renal disease (ESRD). Progress in KT is limited by two major barriers: a) a chronic and burgeoning shortage of transplantable organs and b) the need for chronic immunosuppression following transplantation. Although ground-breaking advances in transplant immunology have improved patient survival and graft durability, a new pathway of innovation is needed in order to overcome current obstacles. Regenerative medicine (RM) holds the potential to shift the paradigm in RRT, through organ bioengineering. Manufactured organs represent a potentially inexhaustible source of transplantable grafts that would bypass the need for immunosuppressive drugs by using autologous cells to repopulate extracellular matrix (ECM) scaffolds. This overview discusses the current status of renal transplantation while reviewing the most promising innovations in RM therapy as applied to RRT.
Collapse
Affiliation(s)
- Jeffrey Rogers
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Ravi Katari
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Sheyna Gifford
- c Annenberg School for Communication & Journalism , University of Southern California , Los Angeles , CA , USA
| | | | - Lauren Edgar
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Marcia R Voigt
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Sean V Murphy
- d Wake Forest Institute for Regenerative Medicine , Winston Salem , NC , USA
| | - Daniel Igel
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Sara Mancone
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Tyler Callese
- b Wake Forest University School of Medicine , Winston Salem , NC , USA
| | - Nicola Colucci
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Majid Mirzazadeh
- e Department of Urology , Wake Forest University , Winston Salem , NC , USA
| | - Andrea Peloso
- f Department of General Surgery , University of Pavia , Pavia , Italy
| | - Joao Paulo Zambon
- d Wake Forest Institute for Regenerative Medicine , Winston Salem , NC , USA
| | - Alan C Farney
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Robert J Stratta
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| | - Giuseppe Orlando
- a Department of Surgery , Wake Forest University , Winston Salem , NC , USA
| |
Collapse
|
32
|
Matz M, Fabritius K, Liu J, Lorkowski C, Brakemeier S, Unterwalder N, Dürr M, Mashreghi MF, Neumayer HH, Budde K. Conversion to Belatacept based regimen does not change T-cell phenotype and function in renal transplantation. Transpl Immunol 2015; 33:176-84. [PMID: 26478531 DOI: 10.1016/j.trim.2015.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/08/2015] [Accepted: 10/13/2015] [Indexed: 11/19/2022]
Abstract
Belatacept offers a new option for renal allograft recipients who are suffering from side effects of calcineurin inhibitors or mTOR inhibitors,which may result in renal and extrarenal benefits.We prospectively performed flow cytometric immunophenotyping with a T-cell panel. In total we were able to fully investigate the immunophenotypic change in 8 patients before and after conversion from calcineurin inhibitor (n = 5) or mTOR inhibitor (n=2) to Belatacept or additional administration (n=1). Cells were analysed pre conversion, 1 month, 3 months, 6 months and 12 months after first Belatacept administration. The percentage of central memory, naïve, effector memory and terminally differentiated effector memory CD4+ and CD4− T-cells was determined. CD28, CD25 and CD69 expression on CD4+ and CD4− T-cells was measured ex vivo and also after 3 days of mitogen stimulation. Intracellular cytokines IFNgamma and IL-2 were measured after polyclonal cellular stimulation. The expression of activation markers and intracellular cytokines as well as the percentage of T-cell subsets did not change significantly during the observation period compared to the time-point pre conversion. Therefore the conversion of calcineurin inhibitor or mTOR inhibitor to Belatacept seems to have no obvious impact on the immunophenotype of T-cells in patients after kidney transplantation.
Collapse
Affiliation(s)
- Mareen Matz
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany.
| | - Katharina Fabritius
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Juan Liu
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Christine Lorkowski
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Susanne Brakemeier
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Nadine Unterwalder
- Labor Berlin GmbH, Immunology Department, Universitätsmedizin Charité Campus Virchow, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Michael Dürr
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | | | - Hans-H Neumayer
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| | - Klemens Budde
- Department of Nephrology, Universitätsmedizin Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
33
|
Meiller TF, Varlotta S, Weikel D. Recognition and Management of Oral Mucosal Injury Caused by Mammalian Target of Rapamycin Inhibitors: A Case Series. Case Rep Oncol 2015; 8:369-77. [PMID: 26464573 PMCID: PMC4592504 DOI: 10.1159/000438747] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The mammalian target of rapamycin inhibitors (mTORIs) everolimus and temsirolimus are approved by the US Food and Drug Administration (FDA) for the treatment of various forms of advanced cancer, and the mTORI sirolimus is approved as an immunosuppressive agent for the prophylaxis of organ rejection in patients receiving renal transplants. The oral lesions associated with mTORI toxicity are distinct from the well-documented chemotherapy- and radiotherapy-induced mucositis, but they may often be misdiagnosed by medical oncologists or transplant physicians, potentially resulting in inappropriate management of this complication. mTORI-associated oral mucosal injury appears to be dose related, and its onset is consistently earlier than conventional mucositis associated with chemotherapy or radiation therapy. Although the lesions appear to resolve within approximately 2 weeks and do not seem to recur as severely with subsequent courses of therapy, the reduction in a patient's quality of life as a result of oral pain that affects the intake of nutritional foods should be taken into consideration. We report three cases that illustrate the complexity involved in the early assessment, referral, and appropriate management of mTORI-associated oral mucosal injury. Corticosteroids appear to be very useful in managing and perhaps preventing these lesions, whereas this approach has never shown efficacy in conventional chemotherapy-related mucositis. Early intervention to reduce the mTORI-associated oral mucosal injury is important to diminish the need for dose alterations of mTORIs and, therefore, to improve patient outcomes.
Collapse
Affiliation(s)
- Timothy F Meiller
- Department of Oncology and Diagnostic Sciences at University of Maryland School of Dentistry, University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical System, Baltimore, Md., USA
| | - Sharon Varlotta
- Department of Oncology and Diagnostic Sciences at University of Maryland School of Dentistry, University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical System, Baltimore, Md., USA
| | - Dianna Weikel
- Department of Oncology and Diagnostic Sciences at University of Maryland School of Dentistry, University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical System, Baltimore, Md., USA
| |
Collapse
|
34
|
Sirolimus plus prednisone for Erdheim-Chester disease: an open-label trial. Blood 2015; 126:1163-71. [PMID: 26041743 DOI: 10.1182/blood-2015-01-620377] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/19/2015] [Indexed: 01/03/2023] Open
Abstract
Erdheim-Chester disease (ECD) is a rare non-Langerhans cell histiocytosis, to whose pathogenesis neoplastic and immune-mediated mechanisms contribute. Mammalian target of rapamycin (mTOR)-inhibitors have antiproliferative and immunosuppressive properties. We tested in this study, the efficacy and safety of the mTOR-inhibitor sirolimus (SRL) plus prednisone (PDN) in patients with ECD. PDN was given initially at 0.75 mg/kg per day, tapered to 5 to 2.5 mg per day by month 6. Target SRL blood levels were 8 to 12 ng/mL. Treatment was continued for at least 24 months in patients who showed disease stabilization or improvement. Ten patients were enrolled; 8 achieved stable disease or objective responses, whereas 2 had disease progression. Responses were mainly observed at the following sites: retroperitoneum in 5/8 patients (62.5%), cardiovascular in 3/4 (75%), bone in 3/9 (33.3%), and central nervous system (CNS) in 1/3 (33.3%). The median follow-up was 29 months (interquartile range, 16.5-74.5); 2 patients died of progressive CNS disease and small-cell lung cancer, respectively. Treatment-related toxicity was mild. Using immunohistochemistry and immunofluorescence on ECD biopsies, we detected expression in foamy histiocytes of the phosphorylated forms of mTOR and of its downstream kinase p70S6K, which indicated mTOR pathway activation. In conclusion, SRL and PDN often induce objective responses or disease stabilization and may represent a valid treatment of ECD. The trial is registered at the Australia-New Zealand Clinical Trial Registry as #ACTRN12613001321730.
Collapse
|
35
|
Everolimus in acute kidney injury in a patient with breast cancer: a case report. J Med Case Rep 2014; 8:386. [PMID: 25420955 PMCID: PMC4364691 DOI: 10.1186/1752-1947-8-386] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 09/22/2014] [Indexed: 11/12/2022] Open
Abstract
Introduction Everolimus, a mammalian target of Rapamycin inhibitor, has recently been approved for the treatment of metastatic estrogen receptor-positive breast cancer, in combination with exemestane at a daily dose of 10mg. In the literature, few cases of acute kidney injury have been reported related to everolimus use, but none of them in a patient with breast cancer as we report here. Our case report of acute kidney injury demonstrates the potential nephrotoxic effects of everolimus therapy, necessitating close monitoring of renal function prior to, during and after discontinuation of the drug. Case presentation We report the first published case of acute kidney injury shortly after initiation of exemestane and everolimus for metastatic breast cancer resistant to letrozole in a 69-year-old Caucasian woman, initially treated for a stage IIB estrogen receptor-positive breast cancer in 1997. Within 2 weeks of therapy, she developed grade 1 to 2 diarrhea, lower extremity edema, lethargy, and anorexia. After 4 weeks of therapy, her blood pressure was 85/59mmHg and she lost 4kg bodyweight. Her serum creatinine was 3.34mg/dL. Everolimus was stopped, and she was hospitalized for rehydration. Her serum creatinine levels peaked at 8.85mg/dL 8 days after treatment discontinuation, with a calculated creatinine clearance of 7mL/minute. Dialysis was not required. A month later, her serum creatinine levels slowly dropped to 2.26mg/dL but did not return to baseline. No re-challenge of everolimus was attempted. Conclusions Extreme vigilance should be used when prescribing everolimus for metastatic breast cancer. Although the exact cause of acute kidney injury in our case is unknown, dehydration must be avoided and renal function closely monitored after initiating therapy. Spontaneous recovery after drug discontinuation is possible.
Collapse
|
36
|
Rodriguez KA, Dodds SG, Strong R, Galvan V, Sharp ZD, Buffenstein R. Divergent tissue and sex effects of rapamycin on the proteasome-chaperone network of old mice. Front Mol Neurosci 2014; 7:83. [PMID: 25414638 PMCID: PMC4220119 DOI: 10.3389/fnmol.2014.00083] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/11/2014] [Indexed: 01/05/2023] Open
Abstract
Rapamycin, an allosteric inhibitor of the mTOR kinase, increases longevity in mice in a sex-specific manner. In contrast to the widely accepted theory that a loss of proteasome activity is detrimental to both life- and healthspan, biochemical studies in vitro reveal that rapamycin inhibits 20S proteasome peptidase activity. We tested if this unexpected finding is also evident after chronic rapamycin treatment in vivo by measuring peptidase activities for both the 26S and 20S proteasome in liver, fat, and brain tissues of old, male and female mice fed encapsulated chow containing 2.24 mg/kg (14 ppm) rapamycin for 6 months. Further we assessed if rapamycin altered expression of the chaperone proteins known to interact with the proteasome-mediated degradation system (PMDS), heat shock factor 1 (HSF1), and the levels of key mTOR pathway proteins. Rapamycin had little effect on liver proteasome activity in either gender, but increased proteasome activity in female brain lysates and lowered its activity in female fat tissue. Rapamycin-induced changes in molecular chaperone levels were also more substantial in tissues from female animals. Furthermore, mTOR pathway proteins showed more significant changes in female tissues compared to those from males. These data show collectively that there are divergent tissue and sex effects of rapamycin on the proteasome-chaperone network and that these may be linked to the disparate effects of rapamycin on males and females. Further our findings suggest that rapamycin induces indirect regulation of the PMDS/heat-shock response through its modulation of the mTOR pathway rather than via direct interactions between rapamycin and the proteasome.
Collapse
Affiliation(s)
- Karl A Rodriguez
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio San Antonio, TX, USA ; Department of Physiology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Sherry G Dodds
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio San Antonio, TX, USA ; Department of Molecular Medicine, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Randy Strong
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio San Antonio, TX, USA ; Department of Pharmacology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Veronica Galvan
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio San Antonio, TX, USA ; Department of Physiology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Z D Sharp
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio San Antonio, TX, USA ; Department of Molecular Medicine, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Rochelle Buffenstein
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio San Antonio, TX, USA ; Department of Physiology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| |
Collapse
|
37
|
Kuik KTH, Trubiano J, Worth LJ, Harun NS, Steinfort D, Johnson D. Pneumocystis jirovecii pneumonia following everolimus treatment of metastatic breast cancer. Med Mycol Case Rep 2014; 6:34-6. [PMID: 25379397 PMCID: PMC4216328 DOI: 10.1016/j.mmcr.2014.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/25/2014] [Accepted: 08/29/2014] [Indexed: 01/22/2023] Open
Abstract
Everolimus is an inhibitor of mammalian target of rapamycin with anti-tumour activity. While everolimus is known to cause drug-induced pneumonitis, it is rarely associated with Pneumocystis jirovecii pneumonia (PJP). We report a patient on everolimus therapy for metastatic breast cancer that developed PJP. Diagnosis was based on clinical features and a quantitative polymerase chain reaction for P. jirovecii DNA. Clinicians should consider PJP as a potential cause of pulmonary infiltrates in patients treated with everolimus.
Collapse
Affiliation(s)
- Kelvin Teck-Hong Kuik
- Department of General Medicine, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Jason Trubiano
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Leon J Worth
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, East Melbourne, Australia ; Department of Medicine, University of Melbourne, Parkville, Australia
| | - Nur-Shirin Harun
- Department of General Medicine, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Daniel Steinfort
- Department of General Medicine, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Douglas Johnson
- Department of General Medicine, Peter MacCallum Cancer Centre, East Melbourne, Australia
| |
Collapse
|
38
|
Su L, Tam N, Deng R, Chen P, Li H, Wu L. Everolimus-based calcineurin-inhibitor sparing regimens for kidney transplant recipients: a systematic review and meta-analysis. Int Urol Nephrol 2014; 46:2035-44. [DOI: 10.1007/s11255-014-0783-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 06/28/2014] [Indexed: 12/31/2022]
|
39
|
Abstract
Morbidity and mortality due to skin cancer is excessively high in renal transplant recipients compared to the general population. This epidemiologic difference is mainly due to the severe immunosuppression that enhances ultraviolet-induced DNA damage and leads to reactivation of potential oncogenic viruses. The most common skin cancer in transplant recipients is squamous cell carcinoma followed by basal cell carcinoma, while in the general population this ratio is reversed. Melanoma and cutaneous lymphoma are relatively rare although they occur more frequently in transplant patients than in the general population. Notably some tumors, such as Kaposi's sarcoma, are seldom encountered in the general population while they are frequently observed in transplant recipients. Local recurrences and visceral spreading are not so uncommon and pose a major issue for quality of life and overall prognosis of these patients. Timely diagnosis is essential and may be challenging, since the accuracy of clinical diagnosis is modest; thus skin biopsy is an essential tool for appropriate management. In this review, we describe the most common types of skin cancer in renal transplant recipients, with a focus on pathogenic issues that account for the different epidemiology and clinical expression of these neoplasms in this population.
Collapse
|