1
|
Plavsa A, Suresh T, Dalal S, Mirea L, Adams RH, Shenoy S, Ngwube A. Comparison of outcomes following subcutaneous or intravenous alemtuzumab administered prior to reduced intensity conditioning for transplantation in pediatric sickle cell disease. Transpl Immunol 2025; 89:102179. [PMID: 39904467 DOI: 10.1016/j.trim.2025.102179] [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: 06/17/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Alemtuzumab-containing conditioning regimens are used for allogeneic hematopoietic stem cell transplantation (HSCT) to reduce acute and chronic graft-versus-host disease (GVHD) and the risk of graft rejection. Alemtuzumab is typically administered intravenously but is often accompanied by infusion-related side effects, including injection site reactions and anaphylaxis. Little is known about the routes of administration and if they differ in safety and efficacy in pediatric patients, especially when used in transplant conditioning. OBJECTIVES To compare adverse effects and efficacy outcomes between intravenous and subcutaneous alemtuzumab administration in pediatric patients with sickle cell disease who have undergone HSCT. STUDY DESIGN A retrospective cohort of 49 pediatric patients with sickle cell disease aged 4-16 years underwent HSCT and received either intravenous or subcutaneous alemtuzumab at St. Louis Children's Hospital or Phoenix Children's Hospital. The incidence of infusion-related reactions, neutrophil and platelet recovery, graft failure, and immune reconstitution were compared. RESULTS We found that subcutaneous alemtuzumab administration elicited fewer infusion-related reactions than intravenously administered drug (p = 0.038). No significant differences in engraftment rates, graft failure rates, infectious complications, acute GVHD, and immune reconstitution were found between the two groups. CONCLUSION Subcutaneous administration of alemtuzumab for children undergoing transplant for sickle cell disease is safe and effective.
Collapse
Affiliation(s)
- Alexandra Plavsa
- Department of Pediatrics, Creighton University School of Medicine, Phoenix, AZ, USA
| | - Tara Suresh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Stuti Dalal
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Lucia Mirea
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Roberta H Adams
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Shalini Shenoy
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Alexander Ngwube
- Department of Pediatrics, Creighton University School of Medicine, Phoenix, AZ, USA; Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA.
| |
Collapse
|
2
|
Korsholm C, Bülow C, Christensen M, Dalhoff K, Feinberg JB, Lund TM, Niemann CU, Petersen TS, Andersen MA. Drug exposure and measurable residual disease in chronic lymphocytic leukemia: a systematic review. Leuk Lymphoma 2025; 66:229-239. [PMID: 39509142 DOI: 10.1080/10428194.2024.2412289] [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: 06/06/2024] [Revised: 09/19/2024] [Accepted: 09/29/2024] [Indexed: 11/15/2024]
Abstract
For fixed-duration therapies against chronic lymphocytic leukemia (CLL), undetectable measurable residual disease (MRD) predicts overall and progression-free survival more accurately than complete remission. For indefinite therapies, MRD status can direct discontinuation of treatment. We systematically reviewed the relationship between antineoplastic drug exposures and undetectable MRD in CLL. Seventeen trials from MEDLINE and EMBASE met the inclusion criteria; four of which evaluated drug exposures in relation to MRD status. Undetectable MRD was associated with higher trough concentrations of ofatumumab and alemtuzumab, as well as increased maximum concentration and area under the plasma concentration curve (AUC) of ibrutinib. One study found an association between high rituximab AUC and undetectable MRD until adjusting for tumor burden. The limited studies, lack of exposure measurements of concomitant drugs, and high heterogeneity in designs limit the results' generalizability. Further research is needed to explore the exposure-MRD relationship and the possibility for therapeutic drug monitoring in CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Neoplasm, Residual
- Antineoplastic Agents/therapeutic use
- Drug Monitoring/methods
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Treatment Outcome
Collapse
Affiliation(s)
- Cathrine Korsholm
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Cille Bülow
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Mikkel Christensen
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kim Dalhoff
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joshua Buron Feinberg
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Trine Meldgaard Lund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Utoft Niemann
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Tonny Studsgaard Petersen
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Asger Andersen
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| |
Collapse
|
3
|
Achini-Gutzwiller FR, Schilham MW, von Asmuth EGJ, Jansen-Hoogendijk AM, Jol-van der Zijde CM, van Tol MJD, Bredius RGM, Güngör T, Lankester AC, Moes DJAR. Exposure-response analysis of alemtuzumab in pediatric allogeneic HSCT for nonmalignant diseases: the ARTIC study. Blood Adv 2023; 7:4462-4474. [PMID: 37285798 PMCID: PMC10440472 DOI: 10.1182/bloodadvances.2022009051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/09/2023] Open
Abstract
Alemtuzumab (anti-CD52 antibody) is frequently prescribed to children with nonmalignant diseases undergoing allogeneic hematopoietic stem cell transplantation (HSCT) to prevent graft failure (GF) and acute graft-versus-host disease (aGVHD). The aim of this multicenter study was the characterization of alemtuzumab population pharmacokinetics to perform a novel model-based exposure-response analysis in 53 children with nonmalignant immunological or hematological disease and a median age of 4.4 years (interquartile range [IQR], 0.8-8.7). The median cumulative alemtuzumab dose was 0.6 mg/kg (IQR, 0.6-1) administered over 2 to 7 days. A 2-compartment population pharmacokinetics model with parallel linear and nonlinear elimination including allometrically scaled bodyweight (median, 17.50 kg; IQR, 8.76-33.00) and lymphocyte count at baseline (mean, 2.24 × 109/L; standard deviation ± 1.87) as significant pharmacokinetic predictors was developed using nonlinear mixed effects modeling. Based on the model-estimated median concentration at day of HSCT (0.77 μg/mL; IQR, 0.33-1.82), patients were grouped into a low- (≤0.77 μg/mL) or high- (>0.77 μg/mL) exposure groups. High alemtuzumab exposure at day of HSCT correlated with delayed CD4+ and CD8+ T-cell reconstitution (P value < .0001) and increased risk of GF (P value = .043). In contrast, alemtuzumab exposure did not significantly influence the incidence of aGVHD grade ≥2, mortality, chimerism at 1 year, viral reactivations, and autoimmunity at a median follow-up of 3.3 years (IQR, 2.5-8.0). In conclusion, this novel population pharmacokinetics model is suitable for individualized intravenous precision dosing to predict alemtuzumab exposure in pediatric allogeneic HSCT for nonmalignant diseases, aiming at the achievement of early T-cell reconstitution and prevention of GF in future prospective studies.
Collapse
Affiliation(s)
- Federica R. Achini-Gutzwiller
- Department of Pediatric Stem Cell Transplantation and Hematology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- Laboratory for Pediatric Immunology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pediatric Stem Cell Transplantation, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco W. Schilham
- Laboratory for Pediatric Immunology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik G. J. von Asmuth
- Laboratory for Pediatric Immunology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Anja M. Jansen-Hoogendijk
- Laboratory for Pediatric Immunology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelia M. Jol-van der Zijde
- Laboratory for Pediatric Immunology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Maarten J. D. van Tol
- Laboratory for Pediatric Immunology, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Robbert G. M. Bredius
- Department of Pediatric Stem Cell Transplantation, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Tayfun Güngör
- Department of Pediatric Stem Cell Transplantation and Hematology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Arjan C. Lankester
- Department of Pediatric Stem Cell Transplantation, Willem-Alexander Children’s Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Jan A. R. Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Koguchi Y, Redmond WL. A Novel Class of On-Treatment Cancer Immunotherapy Biomarker: Trough Levels of Antibody Therapeutics in Peripheral Blood. Immunol Invest 2022; 51:2159-2175. [PMID: 36301695 DOI: 10.1080/08820139.2022.2131570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
While immune checkpoint blockade has revolutionized cancer treatment, unfortunately most patients do not benefit from this treatment. Many pharmacodynamic (PD) studies have revealed essential requirements for successful cancer immunotherapy that may provide insight into how we can improve these agents. Despite enormous efforts focused on interrogating the immune system using different biospecimens (e.g. blood, primary tumor, metastatic tumor, microbiome samples), a variety of technologies (e.g. flow cytometry, bulk and single-cell RNA-sequencing, immunohistochemistry), and wide-ranging disciplines (e.g. pathology, genomics, bioinformatics, immunology, cancer biology, metabolomics, bacteriology), discovery of consistent biomarkers of response have remained elusive. Pharmacokinetics (PK) studies, however, not only provide critical information regarding safe dosing but may also reveal useful biomarkers. For example, recent studies found that trough levels of therapeutic monoclonal antibodies (mAbs) or clearance (CL) of them were associated with clinical outcome, which suggests that trough levels of mAbs may represent a new class of on-treatment cancer immunotherapy biomarker. In this review, we summarize the potential utility of trough levels of mAbs, the mechanism of varying PK, consideration for therapeutic drug monitoring, and assay attributes that will facilitate wider utilization of PK information in conjunction with PD assessments.
Collapse
Affiliation(s)
- Yoshinobu Koguchi
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| |
Collapse
|
5
|
Krajnc N, Bsteh G, Berger T, Mares J, Hartung HP. Monoclonal Antibodies in the Treatment of Relapsing Multiple Sclerosis: an Overview with Emphasis on Pregnancy, Vaccination, and Risk Management. Neurotherapeutics 2022; 19:753-773. [PMID: 35378683 PMCID: PMC8978776 DOI: 10.1007/s13311-022-01224-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 01/10/2023] Open
Abstract
Monoclonal antibodies have become a mainstay in the treatment of patients with relapsing multiple sclerosis (RMS) and provide some benefit to patients with primary progressive MS. They are highly precise by specifically targeting molecules displayed on cells involved in distinct immune mechanisms of MS pathophysiology. They not only differ in the target antigen they recognize but also by the mode of action that generates their therapeutic effect. Natalizumab, an [Formula: see text]4[Formula: see text]1 integrin antagonist, works via binding to cell surface receptors, blocking the interaction with their ligands and, in that way, preventing the migration of leukocytes across the blood-brain barrier. On the other hand, the anti-CD52 monoclonal antibody alemtuzumab and the anti-CD20 monoclonal antibodies rituximab, ocrelizumab, ofatumumab, and ublituximab work via eliminating selected pathogenic cell populations. However, potential adverse effects may be serious and can necessitate treatment discontinuation. Most importantly, those are the risk for (opportunistic) infections, but also secondary autoimmune diseases or malignancies. Monoclonal antibodies also carry the risk of infusion/injection-related reactions, primarily in early phases of treatment. By careful patient selection and monitoring during therapy, the occurrence of these potentially serious adverse effects can be minimized. Monoclonal antibodies are characterized by a relatively long pharmacologic half-life and pharmacodynamic effects, which provides advantages such as permitting infrequent dosing, but also creates disadvantages regarding vaccination and family planning. This review presents an overview of currently available monoclonal antibodies for the treatment of RMS, including their mechanism of action, efficacy and safety profile. Furthermore, we provide practical recommendations for risk management, vaccination, and family planning.
Collapse
Affiliation(s)
- Nik Krajnc
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Gabriel Bsteh
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Jan Mares
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
| | - Hans-Peter Hartung
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic.
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Moorenstrasse 5, 40225, Düsseldorf, Germany.
- Brain and Mind Center, University of Sydney, Sydney, Australia.
| |
Collapse
|
6
|
Immunosuppressive Drugs. ENCYCLOPEDIA OF INFECTION AND IMMUNITY 2022. [PMCID: PMC8987166 DOI: 10.1016/b978-0-12-818731-9.00068-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immunosuppressant is a class of medicines that inhibit or decrease the intensity of the immune response in the body. Most of these medications are used to allow the body less likely to resist a transplanted organ. In solid organ transplantation, immunosuppressive agents are needed for the activation of early-stage immunosuppression, the management of late-stage immunosuppression or for the maintenance of organ rejection. The emergence of novel agents and improvements in immunosuppression regimens after transplantation are significant factors leading to this progress. However, these drugs also increase the risk of infection, cancers and specific adverse side effects specific to each agent in patients particularly in pregnant women and fertility issues. Corona virus disease being hot topic of debate is has given positive outcome to immunosuppressive drugs however need more attention in future. Transplant centers across the world utilize multiple immunosuppression protocols; nevertheless, each patient can require an individually formulated immunosuppression regimen to manage the advantages and possible damage of treatment thus eliminating the likelihood of their primary disease recurrence.
Collapse
|
7
|
Baker D, Asardag AN, Quinn OA, Efimov A, Kang AS. Anti-drug antibodies to antibody-based therapeutics in multiple sclerosis. Hum Antibodies 2021; 29:255-262. [PMID: 34397407 DOI: 10.3233/hab-210453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Multiple sclerosis is the major demyelinating autoimmune disease of the central nervous system. Relapsing MS can be treated by a number of approved monoclonal antibodies that currently target: CD20, CD25 (withdrawn), CD49d and CD52. These all target potentially pathogenic memory B cell subsets and perhaps functionally inhibit pathogenic T cell function. These consist of chimeric, humanized and fully human antibodies. However, despite humanization it is evident that all of these monoclonal antibodies can induce binding and neutralizing antibodies ranging from < 1% to over 80% within a year of treatment. Importantly, it is evident that monitoring these allow prediction of future treatment-failure in some individuals and treatment cessation and switching therefore potentially limiting disease breakthrough and disability accumulation. In response to the COVID-19 pandemic and the need to avoid hospitals, shortened infusion times and extended dose intervals have been implemented, importantly, subcutaneous delivery of alternative treatments or formulations have been developed to allow for home treatment. Therefore, hospital-based and remote monitoring of ADA could therefore be advantageous to optimize patient responses in the future.
Collapse
Affiliation(s)
- David Baker
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - A Nazli Asardag
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Olivia A Quinn
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alex Efimov
- Camstech Limited, Daresbury Laboratory Science and Technology Facilities Council Sci-Tech, Keckwick, Cheshire, UK
| | - Angray S Kang
- Blizard Institute, Barts and London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Centre for Oral Immunobiology and Regenerative Medicine, Dental Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Biolato M, Bianco A, Lucchini M, Gasbarrini A, Mirabella M, Grieco A. The Disease-Modifying Therapies of Relapsing-Remitting Multiple Sclerosis and Liver Injury: A Narrative Review. CNS Drugs 2021; 35:861-880. [PMID: 34319570 PMCID: PMC8354931 DOI: 10.1007/s40263-021-00842-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/28/2021] [Indexed: 02/08/2023]
Abstract
In this narrative review, we analyze pre-registration and post-marketing data concerning hepatotoxicity of all disease-modifying therapies (DMTs) available for the treatment of relapsing-remitting multiple sclerosis, including beta interferon, glatiramer acetate, fingolimod, teriflunomide, dimethyl fumarate, cladribine, natalizumab, alemtuzumab, and ocrelizumab. We review the proposed causal mechanisms described in the literature and we also address issues like use of DMTs in patients with viral hepatitis or liver cirrhosis. Most data emerged in the post-marketing phase by reports to national pharmacovigilance agencies and published case reports or case series. Serious liver adverse events are rare, but exact incidence is largely unknown, as are predictive factors. Unfortunately, none of the DMTs currently available for the treatment of multiple sclerosis is free of potential hepatic toxic effects. Cases of acute liver failure have been reported for beta-interferon, fingolimod, natalizumab, alemtuzumab, and ocrelizumab by different mechanisms (idiosyncratic reaction, autoimmune hepatitis, or viral reactivation). Patients with multiple sclerosis should be informed about possible hepatic side effects of their treatment. Most cases of liver injury are idiosyncratic and unpredictable. The specific monitoring schedule for each DMT has been reviewed and the clinician should be ready to recognize clinical symptoms suggestive for liver injury. Not all DMTs are indicated in cirrhotic patients. For some DMTs, screening for hepatitis B virus and hepatitis C virus is required before starting treatment and a monitoring or antiviral prophylaxis schedule has been established. Beta interferon, glatiramer acetate, natalizumab, and alemtuzumab are relatively contraindicated in autoimmune hepatitis due to the risk of disease exacerbation.
Collapse
Affiliation(s)
- Marco Biolato
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy.
- Institute of Internal Medicine, Catholic University of Sacred Heart, 00168, Rome, Italy.
- Centro di ricerca per la Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, 00168, Rome, Italy.
| | - Assunta Bianco
- Multiple Sclerosis Center, Department of Department of Aging, Neurological, Orthopedic and Head and Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
| | - Matteo Lucchini
- Multiple Sclerosis Center, Department of Department of Aging, Neurological, Orthopedic and Head and Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
- Centro di ricerca per la Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
- Institute of Internal Medicine, Catholic University of Sacred Heart, 00168, Rome, Italy
| | - Massimiliano Mirabella
- Multiple Sclerosis Center, Department of Department of Aging, Neurological, Orthopedic and Head and Neck Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168, Rome, Italy
- Centro di ricerca per la Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Antonio Grieco
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli 8, 00168, Rome, Italy
- Institute of Internal Medicine, Catholic University of Sacred Heart, 00168, Rome, Italy
| |
Collapse
|
9
|
Repetitively Administered Low-Dose Donor Lymphocyte Infusion for Prevention of Relapse after Allogeneic Stem Cell Transplantation in Patients with High-Risk Acute Leukemia. Cancers (Basel) 2021; 13:cancers13112699. [PMID: 34070786 PMCID: PMC8198731 DOI: 10.3390/cancers13112699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Patients with high-risk acute leukemia (AL) have a high risk of relapse after allogeneic stem cell transplantation (allo-SCT). Prophylactic donor lymphocyte infusion (pro-DLI) has shown significant efficacy in reducing relapse rate but its widespread application has been limited due to concerns of graft versus host disease (GVHD) development. In the present study, we tested the safety and efficacy of a novel method of pro-DLI based on repetitive administration of low lymphocyte doses. Low-dose pro-DLI was administered every two months for at least 3-years. Forty-four high-risk AL patients with a median age of 53-years who underwent allo-SCT from a matched-sibling (38pts) or a matched-unrelated donor (6pts) received pro-DLI. Acute and chronic-GVHD developed in 14% and 12% of patients. With a median follow-up of 44-months, the cumulative incidence of relapse and non-relapse mortality was 12% and 13%, while the overall survival was 78%. Prolonged up to 3-years low-dose pro-DLI administered every two months is safe and effective in reducing relapse rate in patients with high-risk AL. The low-dose repetitive administration DLI strategy reduced the risk of DLI-mediated GVHD, while the prolonged repeated administration helped in preventing relapse, possibly by inducing a sustained and prolonged immunological pressure on residual leukemic cells. Abstract Background: Patients with high-risk acute leukemia have a high risk of relapse after allogeneic stem cell transplantation (allo-SCT). In an effort to reduce the relapse rate, various therapeutic methods have been implemented into clinical practice. Among them, prophylactic donor lymphocyte infusion (pro-DLI) has shown significant efficacy. However, the widespread application of pro-DLI has been restricted mostly due to concerns regarding the development of graft versus host disease (GVHD). In the present study, we tested the safety and efficacy of a novel method of prophylactic-DLI based by repetitive administration of low lymphocyte doses. Methods: DLI was administered to patients with high-risk acute leukemia at a dose of 2 × 106/kg CD3-positive cells. DLI at the same dose was repeated every two months for at least 36 months post-allo-SCT, or until relapse or any clinical or laboratory feature suggested GVHD, whichever occurred first. Forty-four patients with a median age of 53 years (range 20–67) who underwent allo-SCT between 2011 and 2020 were included in our study. Thirty-three patients with high-risk acute myeloid leukemia (AML) and 11 with high-risk acute lymphoblastic leukemia (ALL) after allo-SCT from a matched sibling (MSD, no = 38 pts) or a matched-unrelated donor (MUD, no = 6 pts) received pro-DLI. Twenty-three patients were in CR1, all with unfavorable genetic features; 12 patients were in CR2 or beyond; and 9 patients had refractory disease at the time of transplant. Ten out of 23 patients in CR1 had detectable minimal residual disease (MRD) at the time of allo-SCT. Disease risk index (DRI) was high and intermediate in 21 and 23 patients, respectively. Conditioning was myeloablative (MAC) in 36 and reduced intensity (RIC) in 8 patients, while GVHD prophylaxis consisted of cyclosporine-A in combination with low-dose alemtuzumab in 39 patients or with low-dose MTX in 5 patients, respectively. Results: Thirty-five patients completed the scheduled treatment and received a median of 8 DLI doses (range 1–35). Fifteen out of 35 patients received all planned doses, while DLI was discontinued in 20 patients. Reasons for discontinuation included GVHD development in nine, donor unavailability in seven, disease relapse in three, and secondary malignancy in one patient, respectively. Nine patients were still on treatment with DLI, and they received a median of four (range 2–12) doses. Fourteen percent of patients developed transient grade-II acute GVHD while 12% developed chronic GVHD post-DLI administration. Acute GVHD was managed successfully with short course steroids, and four out of five patients with cGVHD were disease-free and off immunosuppression. With a median follow-up of 44 months (range 8–120), relapse-free (RFS) and overall survival (OS) were 74%, (95% CI, 54–87%) and 78%, (95% CI, 58–89%) respectively, while the cumulative incidence of non-relapse mortality (NRM) was 13% (95% CI, 4–28%). The cumulative incidence of relapse in patients with intermediate and high DRI is 7% and 15%, respectively. Conclusion: Prolonged—up to three years—low-dose pro-DLI administered every two months is safe and effective in reducing relapse rate in patients with high-risk acute leukemia. The low-dose repetitive administration DLI strategy reduced the risk of DLI-mediated GVHD, while the prolonged repeated administration helped in preventing relapse, possibly by inducing a sustained and prolonged immunological pressure on residual leukemic cells. This novel strategy deserves testing in larger cohort of patients with high-risk acute leukemia.
Collapse
|
10
|
Manibalan S, Thirukumaran K, Varshni M, Shobana A, Achary A. Report on biopharmaceutical profile of recent biotherapeutics and insilco docking studies on target bindings of known aptamer biotherapeutics. Biotechnol Genet Eng Rev 2021; 36:57-80. [PMID: 33393433 DOI: 10.1080/02648725.2020.1858395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Accumulated Toxicity, disease recurrence and drug resistivity problems have been observed due to the synthetic and semisynthetic therapeutic practices, which alternatively led to focus on Bio-therapeutics production than xenobiotics. Quick plasma clearance and high potency are the reasons for trending research with huge pharma market of numerous Bio-therapeutics than ever before. Researchers proved that most of the nano and micro Bio-therapeutics have multiple beneficial therapeutic effects. We have analyzed the past, and present scenario of some notable clinically approved Bio-therapeutics to identify the future formulation needs with advanced techniques. Protein-related drugs are the foremost Bio-therapeutics such as antibodies, enzymes, and short, fragmented polypeptides show aggregation properties during storage, naked peptide moieties are resisted by the polar cell membrane, and also the antidrug antibodies were reported. Even though Nucleic acid nano-bodies are excellent target binders than proteins, they had only a few minutes of half-life. Maintaining homogeneousness upon storage of Bio-therapeutics is still a significant challenge in industrial-scale formulation. Notably, plant systems are identified as most useful cost-effective hosts to produce human enzymes than animal systems without any possible viral loads. Irrespective of numerous advancements in routes of administration and additives, subcutaneous is still a golden one to achieve better dynamics. Additionally, the interactions and effective bonds made by each class of well-known aptamer biotherapeutics which are considered as future drugs were studied.
Collapse
Affiliation(s)
- Subramaniyan Manibalan
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Kandasamy Thirukumaran
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Mathimaran Varshni
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Ayyasamy Shobana
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Anant Achary
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| |
Collapse
|
11
|
Woodley WD, Yue W, Morel DR, Lainesse A, Pettis RJ, Bolick NG. Clinical Evaluation of an Investigational 5 mL Wearable Injector in Healthy Human Subjects. Clin Transl Sci 2020; 14:859-869. [PMID: 33278331 PMCID: PMC8212760 DOI: 10.1111/cts.12946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/09/2020] [Indexed: 11/26/2022] Open
Abstract
An investigational wearable injector (WI), the BD Libertas Wearable Injector (BD Libertas is a trademark of Becton, Dickinson and Company), was evaluated in an early feasibility clinical study for functional performance, tissue effects, subject tolerability, and acceptability of 5 mL, non‐Newtonian ~ 8 cP subcutaneous placebo injections in 52 healthy adult subjects of 2 age groups (18–64 years and ≥ 65 years). Randomized WI subcutaneous injections (n = 208, 4/subject) were delivered to the right and left abdomen and thigh of each subject, 50% (1 thigh and 1 abdomen) with a defined movement sequence during injection. Injector functional performance was documented. Deposition was qualified and quantified with ultrasound. Tissue effects and tolerability (pain) were monitored through 24 hours with corresponding acceptability questionnaires administered through 72 hours. WI (n = 205) automatically inserted the needle, delivered 5 mL ± 5% in 5.42 minutes (SD 0.74) and retracted. Depots were entirely (93.2%) or predominantly (5.4%) localized within the target subcutaneous tissue. Slight to moderate wheals (63.9%) and erythema (75.1%) were observed with ≥ 50% resolution within 30–60 minutes. Subject pain (100 mm Visual Analog Scale) peaked mid‐injection (mean 9.1 mm, SD 13.4) and rapidly resolved within 30 minutes (mean 0.4 mm, SD 2.6). Subjects’ peak pain (≥ 90.2%), injection site appearance (≥ 92.2%) and injector wear, size, and removal (≥ 92.1%) were acceptable (Likert responses) with 100% likely to use the injector if prescribed. Injection site preference was divided between none (46%), abdomen (25%), or thigh (26.9%). The investigational WI successfully delivered 5 mL viscous subcutaneous injections. Tissue effects and pain were transient, well‐tolerated and acceptable. Neither injection site, movement or subject age affected injector functional performance or subject pain and acceptability.
Collapse
Affiliation(s)
- Wendy D Woodley
- BD Technologies & Innovation, Research Triangle Park, North Carolina, USA
| | - Wen Yue
- BD, Franklin Lakes, New Jersey, USA
| | | | | | - Ronald J Pettis
- BD Technologies & Innovation, Research Triangle Park, North Carolina, USA
| | - Natasha G Bolick
- BD Technologies & Innovation, Research Triangle Park, North Carolina, USA
| |
Collapse
|
12
|
Immune reconstitution, glomerulonephritis, and successful treatment with rituximab. Clin Nephrol Case Stud 2020; 8:67-71. [PMID: 32905274 PMCID: PMC7469242 DOI: 10.5414/cncs110061] [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: 09/25/2019] [Accepted: 05/18/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Alemtuzumab can induce secondary autoimmunity affecting multiple organs. While kidney involvement is uncommon, it can be associated with devastating forms of glomerulonephritis (GN). CASE PRESENTATION A 32-year-old African American woman presented with hypertension, proteinuria, and progressive renal failure. Her medical history was remarkable for secondary progressive multiple sclerosis (SPMS). She had received her first induction dose of alemtuzumab 1 year prior to presentation. Upon evaluation, she had scanning speech, multidirectional nystagmus, and mild edema. Her serum creatinine was 2 mg/dL. Urine studies revealed proteinuria and microscopic hematuria. Her serologic tests were positive for c-antineutrophil cytoplasmic antibodies (> 1 : 640). In addition, she was found to have new-onset severe thyroid dysfunction with antibodies against thyroglobulin and thyroid peroxidase. Kidney biopsy was diagnostic for pauci-immune crescentic GN. The patient was treated with methylprednisolone and rituximab with subsequent renal, thyroid, and neurological recovery. CONCLUSION This is an atypical case of GN following therapy with alemtuzumab. We hypothesize that immune reconstitution may be a potential mechanism. Alemtuzumab is a new treatment for SPMS that can be associated with GN. Practice guidelines should address the management of its renal complications.
Collapse
|
13
|
Rolla S, Maglione A, De Mercanti SF, Clerico M. The Meaning of Immune Reconstitution after Alemtuzumab Therapy in Multiple Sclerosis. Cells 2020; 9:E1396. [PMID: 32503344 PMCID: PMC7348777 DOI: 10.3390/cells9061396] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Alemtuzumab is a monoclonal antibody that binds to CD52, a protein present on the surface of mature lymphocytes, but not on the stem cells from which these lymphocytes are derived. It is currently used as an immune reconstitution therapy in patients with relapsing-remitting multiple sclerosis. Alemtuzumab treatment is an intermittent infusion that induces long-term remission of Multiple Sclerosis also in the treatment-free period. After the robust T and B cell depletion induced by alemtuzumab, the immune system undergoes radical changes during its reconstitution. In this review, we will discuss the current knowledge on the reconstitution of the lymphocyte repertoire after alemtuzumab treatment and how it could affect the development of side effects, which led to its temporary suspension by the European Medical Agency.
Collapse
Affiliation(s)
- Simona Rolla
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano (TO), Italy; (A.M.); (S.F.D.M.); (M.C.)
| | | | | | | |
Collapse
|
14
|
|
15
|
Loeff FC, van Egmond EH, Moes DJ, Wijnands C, Von Dem Borne PA, Veelken H, Falkenburg JF, Jedema I, Halkes CJ. Impact of alemtuzumab pharmacokinetics on T-cell dynamics, graft-versus-host disease and viral reactivation in patients receiving allogeneic stem cell transplantation with an alemtuzumab-based T-cell-depleted graft. Transpl Immunol 2019; 57:101209. [DOI: 10.1016/j.trim.2019.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 10/26/2022]
|
16
|
Roos JCP, Moran C, Chatterjee VK, Jones J, Coles A, Murthy R. Immune reconstitution after alemtuzumab therapy for multiple sclerosis triggering Graves' orbitopathy: a case series. Eye (Lond) 2019; 33:223-229. [PMID: 30498266 PMCID: PMC6367353 DOI: 10.1038/s41433-018-0282-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 11/06/2018] [Indexed: 11/08/2022] Open
Abstract
Alemtuzumab-a monoclonal antibody targeting the CD52 glycoprotein expressed by most mature leucocytes-effectively decreases relapse rate and disability progression in early, relapsing-remitting multiple sclerosis (MS). However, secondary autoimmune disorders complicate therapy in nearly 50% of treated patients, with Graves' disease being the most common. Rarely, thyroid eye disease (TED) ensues; only seven such cases have been reported. Our aim was to analyse the largest series of MS patients developing thyroid eye disease after alemtuzumab treatment. We performed a retrospective chart review of MS patients treated with alemtuzumab (1995-2018) and subsequently identified by their treating physicians as having developed TED and referred to our ophthalmology service. As an original trial centre for alemtuzumab, our hospital has treated approximately 162 MS patients with this novel therapy. In total, 71 (44%) developed thyroid dysfunction, most of whom (87%) developed Graves' disease, with ten (16%) referred for ophthalmological evaluation. Two developed active orbitopathy following radioiodine treatment; one occurred after cessation of anti-thyroid drug treatment. Three developed sight-threatening disease requiring systemic immunosuppression, with one refractory to multiple immunosuppressants. The remaining patients were treated conservatively. TSH-receptor antibody (TRAb) levels were significantly raised in all cases, when ascertained. We report sight-threatening as well as mild TED in MS patients after treatment with alemtuzumab. Endocrine instability, radioiodine treatment and positive TRAb are all likely risk factors. The data support at least 6-monthly biochemical and clinical assessment with a low threshold for referral to an ophthalmologist, particularly for those with higher TRAb levels who may be at greater risk of orbitopathy.
Collapse
Affiliation(s)
- Jonathan C P Roos
- Thyroid Eye Disease Service, Department of Ophthalmology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Carla Moran
- Wellcome-MRC Institute of Metabolic Science, Cambridge, UK
| | | | - Joanne Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Alasdair Coles
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Rachna Murthy
- Thyroid Eye Disease Service, Department of Ophthalmology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
Immunoassays for Measuring Serum Concentrations of Monoclonal Antibodies and Anti-biopharmaceutical Antibodies in Patients. Ther Drug Monit 2018; 39:316-321. [PMID: 28570370 DOI: 10.1097/ftd.0000000000000419] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Monoclonal antibodies (mAbs) may be used as biopharmaceuticals to treat various diseases, ranging from oncology to inflammatory and cardiovascular affections. Trustworthy analytical methods are necessary to study their pharmacokinetics, both during their development and in post-marketing studies. Because biopharmaceuticals are macromolecules, ligand-binding assays (both immunoassays and bioassays) are methods of choice to measure their concentrations. Immunoassays are based on the capture of biopharmaceuticals by their target, which may be a circulating or membrane antigen or by an antibody recognizing their structure. Bioassays measure the activity of the biopharmaceutical in a specific in vitro test. A number of techniques have been reported, but their limits of detection and quantification vary widely. Anti-drug antibodies (ADA) against biopharmaceuticals are often formed and sometimes interfere with clinical efficacy. Accurate and reliable detection of ADA is therefore necessary. Binding of ADA is dependent on affinity and avidity, which makes quantification challenging. In this review, we discuss the benefits and limitations of each method to determine mAb levels and carefully compare ADA assays.
Collapse
|
18
|
Khambalia HA, Alexander MY, Nirmalan M, Weston R, Pemberton P, Moinuddin Z, Summers A, van Dellen D, Augustine T. Links between a biomarker profile, cold ischaemic time and clinical outcome following simultaneous pancreas and kidney transplantation. Cytokine 2018; 105:8-16. [PMID: 29428804 DOI: 10.1016/j.cyto.2018.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/27/2017] [Accepted: 01/08/2018] [Indexed: 01/18/2023]
Abstract
In sepsis, trauma and major surgery, where an explicit physiological insult leads to a significant systemic inflammatory response, the acute evolution of biomarkers have been delineated. In these settings, Interleukin (IL) -6 and TNF-α are often the first pro-inflammatory markers to rise, stimulating production of acute phase proteins followed by peaks in anti-inflammatory markers. Patients undergoing SPKT as a result of diabetic complications already have an inflammatory phenotype as a result of uraemia and glycaemia. How this inflammatory response is affected further by the trauma of major transplant surgery and how this may impact on graft survival is unknown, despite the recognised pro-inflammatory cytokines' detrimental effects on islet cell function. The aim of the study was to determine the evolution of biomarkers in omentum and serum in the peri-operative period following SPKT. The biochemical findings were correlated to clinical outcomes. Two omental biopsies were taken (at the beginning and end of surgery) and measured for CD68+ and CD206+ antibodies (M1 and M2 macrophages respectively). Serum was measured within the first 72 h post-SPKT for pro- and anti-inflammatory cytokines (IL -6, -10 and TNF-α), inflammatory markers (WCC and CRP) and endocrine markers (insulin, C-peptide, glucagon and resistin). 46 patients were recruited to the study. Levels of M1 (CD68+) and M2 (CD206+) macrophages were significantly raised at the end of surgery compared to the beginning (p = 0.003 and p < 0.001 respectively). Levels of C-peptide, insulin and glucagon were significantly raised 30 min post pancreas perfusion compared to baseline and were also significantly negatively related to prolonged cold ischaemic time (CIT) (p < 0.05). CRP levels correlated significantly with the Post-Operative Morbidity Survey (p < 0.05). The temporal inflammatory marker signature after SPKT is comparable to the pattern observed following other physiological insults. Unique to this study, we find that CIT is significantly related to early pancreatic endocrine function. In addition, this study suggests a predictive value of CRP in peri-operative morbidity following SPKT.
Collapse
Affiliation(s)
- Hussein A Khambalia
- Department of Transplantation, Manchester Foundations Hospitals NHS Foundation Trust, Manchester Royal Infirmary, Manchester, United Kingdom.
| | - M Yvonne Alexander
- Cardiovascular Research Inst, University of Manchester, Manchester Academic Health Science Centre, United Kingdom; Healthcare Science Research Institute, Manchester Metropolitan University, Manchester, United Kingdom
| | - Mahesan Nirmalan
- Department of Transplantation, Manchester Foundations Hospitals NHS Foundation Trust, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Ria Weston
- Cardiovascular Research Inst, University of Manchester, Manchester Academic Health Science Centre, United Kingdom
| | - Phillip Pemberton
- Department of Transplantation, Manchester Foundations Hospitals NHS Foundation Trust, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Zia Moinuddin
- Department of Transplantation, Manchester Foundations Hospitals NHS Foundation Trust, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Angela Summers
- Department of Transplantation, Manchester Foundations Hospitals NHS Foundation Trust, Manchester Royal Infirmary, Manchester, United Kingdom
| | - David van Dellen
- Department of Transplantation, Manchester Foundations Hospitals NHS Foundation Trust, Manchester Royal Infirmary, Manchester, United Kingdom
| | - Titus Augustine
- Department of Transplantation, Manchester Foundations Hospitals NHS Foundation Trust, Manchester Royal Infirmary, Manchester, United Kingdom
| |
Collapse
|
19
|
van der Zwan M, Baan CC, van Gelder T, Hesselink DA. Review of the Clinical Pharmacokinetics and Pharmacodynamics of Alemtuzumab and Its Use in Kidney Transplantation. Clin Pharmacokinet 2018; 57:191-207. [PMID: 28669130 PMCID: PMC5784003 DOI: 10.1007/s40262-017-0573-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alemtuzumab is a humanized monoclonal antibody against CD52 and causes depletion of T and B lymphocytes, monocytes, and NK cells. Alemtuzumab is registered for the treatment of multiple sclerosis (MS) and is also used in chronic lymphocytic leukemia (CLL). Alemtuzumab is used off-label in kidney transplantation as induction and anti-rejection therapy. The objective of this review is to present a review of the pharmacokinetics, pharmacodynamics, and use of alemtuzumab in kidney transplantation. A systematic literature search was conducted using Ovid Medline, Embase, and Cochrane Central Register of controlled trials. No pharmacokinetic or dose-finding studies of alemtuzumab have been performed in kidney transplantation. Although such studies were conducted in patients with CLL and MS, these findings cannot be directly extrapolated to transplant recipients, because CLL patients have a much higher load of CD52-positive cells and, therefore, target-mediated clearance will differ between these two indications. Alemtuzumab used as induction therapy in kidney transplantation results in a lower incidence of acute rejection compared to basiliximab therapy and comparable results as compared with rabbit anti-thymocyte globulin (rATG). Alemtuzumab used as anti-rejection therapy results in a comparable graft survival rate compared with rATG, although infusion-related side effects appear to be less. There is a need for pharmacokinetic and dose-finding studies of alemtuzumab in kidney transplant recipients to establish the optimal balance between efficacy and toxicity. Furthermore, randomized controlled trials with sufficient follow-up are necessary to provide further evidence for the treatment of severe kidney transplant rejection.
Collapse
Affiliation(s)
- Marieke van der Zwan
- Division of Nephrology and Kidney Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Room NA523, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Carla C Baan
- Division of Nephrology and Kidney Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Room NA523, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Teun van Gelder
- Division of Nephrology and Kidney Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Room NA523, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
- Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dennis A Hesselink
- Division of Nephrology and Kidney Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Room NA523, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW We aimed to produce a comprehensive update on clinical and biological data regarding two rare lymphoid neoplasms, B and T prolymphocytic leukemias, and assess therapeutic management in the light of new molecular insights and the advent of targeted therapies. RECENT FINDINGS B cell prolymphocytic leukemia (B-PLL) diagnosis remains challenging in the absence of clear immunophenotypic or cytogenetic signature and overlap with mantle cell lymphoma. New molecular defects have been identified in T cell prolymphocytic leukemia (T-PLL), especially in the JAK STAT pathway. Like in chronic lymphocytic leukemia (CLL), B-PLL treatment depends on the presence of TP53 dysfunction. In T-PLL, alemtuzumab still remains the standard of care. Allogeneic transplantation is the only curable option. Thanks to reduced intensity conditioning regimens, it has become accessible to a larger number of patients. PLL prognosis remains poor with conventional therapies. However, great advances in the understanding of both T- and B-PLL pathogenesis lead to promising new therapeutic agents.
Collapse
|
21
|
Assouline S, Buccheri V, Delmer A, Gaidano G, Trneny M, Berthillon N, Brewster M, Catalani O, Li S, McIntyre C, Sayyed P, Badoux X. Pharmacokinetics, safety, and efficacy of subcutaneous versus intravenous rituximab plus chemotherapy as treatment for chronic lymphocytic leukaemia (SAWYER): a phase 1b, open-label, randomised controlled non-inferiority trial. LANCET HAEMATOLOGY 2017; 3:e128-38. [PMID: 26947201 DOI: 10.1016/s2352-3026(16)00004-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/22/2015] [Accepted: 01/04/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Part one of the two-part SAWYER study predicted that subcutaneous rituximab 1600 mg would achieve trough serum concentrations that were non-inferior to those achieved with intravenous rituximab 500 mg/m(2) in patients with chronic lymphocytic leukaemia. In part two of the study, we aimed to confirm the pharmacokinetic non-inferiority of subcutaneous rituximab, and investigate its safety and efficacy. METHODS We did this phase 1b, open-label, randomised controlled non-inferiority study at 68 centres in 19 countries in Europe, North America, South America, and Australasia. Patients aged 18 years or older with untreated chronic lymphocytic leukaemia were randomly assigned, via an interactive voice-response system with a permuted block randomisation scheme (block size of ten), to receive subcutaneous rituximab 1600 mg or intravenous rituximab 500 mg/m(2) plus fludarabine and cyclophosphamide every 4 weeks for up to six cycles. In cycle one, all patients received intravenous rituximab 375 mg/m(2). Randomisation was stratified by Binet stage and fludarabine and cyclophosphamide administration route (oral vs intravenous). Study investigators and patients were not masked to group allocation, but allocation was concealed from the statistician, clinical scientist, and clinical pharmacologist. The primary endpoint was trough serum concentration at cycle five, with a non-inferiority margin of 0·8 for the adjusted geometric mean ratio of the subcutaneous to the intravenous dose. We did the primary analysis in patients in the intention-to-treat population with complete pharmacokinetic data (pharmacokinetic population). This trial is registered with ClinicalTrials.gov, number NCT01292603, and is ongoing, although the treatment stage is now complete. FINDINGS Between Aug 20, 2012, and June 17, 2013, we randomly assigned 176 patients to receive subcutaneous rituximab (n=88) or intravenous rituximab (n=88); 134 (76%) patients comprised the pharmacokinetic population. As of May 7, 2014, median follow-up was 13·9 months (IQR 11·9-16·0) for patients in the subcutaneous group and 14·1 months (11·6-16·5) for patients in the intravenous group. At cycle five, the geometric mean trough serum concentration in patients given subcutaneous rituximab was non-inferior to that in patients given intravenous rituximab (97·5 μg/mL vs 61·5 μg/mL), with an adjusted geometric mean ratio of 1·53 (90% CI 1·27-1·85). In the safety analysis, the proportion of patients reporting adverse events was similar between the subcutaneous and intravenous groups (all grades: 82 [96%] of 85 patients and 81 [91%] of 89 patients; serious adverse events: 25 [29%] and 29 [33%] patients; grade ≥3: 59 [69%] and 63 [71%] patients, respectively). The most common adverse event of grade 3 or higher was neutropenia (48 [56%] patients in the subcutaneous group and 46 [52%] patients in the intravenous group); the most common serious adverse event was febrile neutropenia (n=9 [11%] and n=4 [4%], respectively). We recorded administration-related reactions in 37 (44%) patients given subcutaneous rituximab and 40 (45%) patients given the intravenous dose, with differences between administration routes for injection-site erythema (n=10 [12%] and n=0, respectively) and nausea (n=2 [2%] and n=11 [12%], respectively). More patients reported local cutaneous reactions after subcutaneous rituximab (n=36 [42%]) than after intravenous rituximab (n=2 [2%]); most of these reactions were grade 1 or 2. INTERPRETATION When combined with fludarabine and cyclophosphamide, subcutaneous rituximab 1600 mg achieved trough serum concentrations that were pharmacokinetically non-inferior to those achieved with intravenous rituximab 500 mg/m(2), with a similar safety and efficacy profile between the two groups. Treatment with subcutaneous rituximab should allow patients with chronic lymphocytic leukaemia to receive clinical benefit from the drug via a more convenient delivery method than the intravenous route, and might also be used in combination regimens with approved and emerging oral regimens. FUNDING F Hoffmann-La Roche.
Collapse
Affiliation(s)
- Sarit Assouline
- Jewish General Hospital, McGill University, Montréal, QC, Canada.
| | - Valeria Buccheri
- Hematology Division-Clinics Hospital, University of São Paulo, São Paulo, Brazil
| | - Alain Delmer
- Department of Haematology, Hôpital Robert Debré, Reims, France
| | - Gianluca Gaidano
- Division of Haematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Marek Trneny
- Charles University, General Hospital Prague, Prague, Czech Republic
| | | | - Michael Brewster
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Welwyn, UK
| | | | - Sai Li
- F Hoffmann-La Roche, Basel, Switzerland
| | - Christine McIntyre
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Welwyn, UK
| | | | - Xavier Badoux
- Department of Haematology, St George Hospital, Sydney, NSW, Australia
| |
Collapse
|
22
|
Schetelig J, de Wreede LC, Andersen NS, Moreno C, van Gelder M, Vitek A, Karas M, Michallet M, Machaczka M, Gramatzki M, Beelen D, Finke J, Delgado J, Volin L, Passweg J, Dreger P, Schaap N, Wagner E, Henseler A, van Biezen A, Bornhäuser M, Iacobelli S, Putter H, Schönland SO, Kröger N. Centre characteristics and procedure-related factors have an impact on outcomes of allogeneic transplantation for patients with CLL: a retrospective analysis from the European Society for Blood and Marrow Transplantation (EBMT). Br J Haematol 2017; 178:521-533. [DOI: 10.1111/bjh.14791] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/20/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Johannes Schetelig
- Medical Department I; University Hospital of the Technical University Dresden; Dresden Germany
- DKMS Clinical Trials Unit; Dresden Germany
| | - Liesbeth C. de Wreede
- DKMS Clinical Trials Unit; Dresden Germany
- Department of Medical Statistics & Bioinformatics; Leiden University Medical Centre; Leiden The Netherlands
| | - Niels S. Andersen
- BMT Unit Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | - Carol Moreno
- Hematologia; Hospital de la Santa Creu i Sant Pau; Barcelona Spain
| | | | - Antonin Vitek
- Department of Haematology; Institute of Haematology and Blood Transfusion; Prague Czech Republic
| | - Michal Karas
- Department of Haematology/Oncology; Charles University Hospital; Pilsen Czech Republic
| | | | - Maciej Machaczka
- Haematology Centre Karolinska and Department of Medicine at Huddinge; Karolinska Institutet; Stockholm Sweden
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy; University Hospital Schleswig-Holstein; Kiel Germany
| | - Dietrich Beelen
- Department of Bone Marrow Transplantation; University Hospital; Essen Germany
| | - Jürgen Finke
- Department of Medicine - Haematology, Oncology; University of Freiburg; Freiburg Germany
| | - Julio Delgado
- Institute of Haematology & Oncology; Department of Haematology; Hospital Clinic; Barcelona Spain
| | - Liisa Volin
- Stem Cell Transplantation Unit; Helsinki University Hospital Comprehensive Cancer Centre; Helsinki Finland
| | - Jakob Passweg
- Department for Haematology; University Hospital; Basel Switzerland
| | - Peter Dreger
- Medizinische Klinik u. Poliklinik V; University of Heidelberg; Heidelberg Germany
| | | | - Eva Wagner
- University Medical Centre Mainz; Mainz Germany
| | - Anja Henseler
- Department of Medical Statistics & Bioinformatics; Leiden University Medical Centre; Leiden The Netherlands
| | - Anja van Biezen
- Department of Medical Statistics & Bioinformatics; Leiden University Medical Centre; Leiden The Netherlands
| | - Martin Bornhäuser
- Medical Department I; University Hospital of the Technical University Dresden; Dresden Germany
| | | | - Hein Putter
- Department of Medical Statistics & Bioinformatics; Leiden University Medical Centre; Leiden The Netherlands
| | - Stefan O. Schönland
- Department of Haematology; Institute of Haematology and Blood Transfusion; Prague Czech Republic
| | - Nicolaus Kröger
- Bone Marrow Transplantation Centre; University Hospital Eppendorf; Hamburg Germany
| | | |
Collapse
|
23
|
Iwata Y, Harada A, Hara T, Kubo C, Inoue T, Tabo M, Ploix C, Manigold T, Hinton H, Mishima M. Is an in vitro whole blood cytokine assay useful to detect the potential risk of severe infusion reaction of monoclonal antibody pharmaceuticals? J Toxicol Sci 2017; 41:523-31. [PMID: 27432238 DOI: 10.2131/jts.41.523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
After the life-threatening cytokine release syndrome (CRS) occurred in the clinical study of the anti-CD28 monoclonal antibody (mAb) TGN1412, in vitro cytokine release assays using human blood cells have been proposed for non-clinical evaluation of the potential risk of CRS. Two basic assay formats are frequently used: human peripheral blood mononuclear cells (PBMC) with immobilized mAbs, and whole blood with aqueous mAbs. However, the suitability of the whole blood cytokine assay (WBCA) has been questioned, because an unrealistically large sample size would be required to detect the potential risk of CRS induced by TGN1412, which has low sensitivity. We performed a WBCA using peripheral blood obtained from 68 healthy volunteers to compare two high risk mAbs, the TGN1412 analogue anti-CD28 superagonistic mAb (CD28SA) and the FcγR-mediated alemutuzumab, with a low risk mAb, panitumumab. Based on the cytokine measurements in this study, the sample size required to detect a statistically significant increase in cytokines with 90% power and 5% significance was determined to be n = 9 for CD28SA and n = 5 for alemtuzumab. The most sensitive marker was IL-8. The results suggest that WBCA is a practical test design that can warn of the potential risk of FcγR-mediated alemtuzumab and T-cell activating CD28SA but, because there was apparently a lower response to CD28SA, it cannot be used as a risk-ranking tool. WBCA is suggested to be a helpful tool for identifying potential FcγR-mediated hazards, but further mechanistic understanding of the response to CD28SA is necessary before applying it to T cell-stimulating mAbs.
Collapse
|
24
|
Abstract
Monoclonal antibodies (mAbs) and fusion proteins with an Fc portion of immunoglobulin G (IgG) are emblematic of the remarkable expansion of biopharmaceuticals. Despite their biological origin, these products display an interindividual variability in their efficacy and/or side effects, which must be taken into consideration. Biological monitoring allowing for adapted prescription and dose adjustments may lead to therapeutic optimization and limitation of the high costs of these drugs. Herein, we review the biological theranostic of mAbs and Fc fusion proteins, including pre-treatment analyses, monitoring of efficacy, therapeutic drug monitoring, and monitoring of side effects. Supported by concrete evidence, a specific interest is given to individualised therapeutic monitoring that combines intention to treat, biomarkers of efficacy and adaptation of serum concentrations.
Collapse
Affiliation(s)
- Benjamin Chaigne
- Université Paris Descartes, Faculté de Médecine, Service de Médecine Interne, Centre de Référence Pour les Vascularites Nécrosantes et la Sclérodermie Systémique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Paris, France
| | - Hervé Watier
- CHRU de Tours, Laboratoire d'Immunologie, France; Université François-Rabelais de Tours, France; CNRS, UMR 7292, Tours, France.
| |
Collapse
|
25
|
Varghese AM, Howard DR, Pocock C, Rawstron AC, Follows G, McCarthy H, Dearden C, Fegan C, Milligan D, Smith AF, Gregory W, Hillmen P. Eradication of minimal residual disease improves overall and progression-free survival in patients with chronic lymphocytic leukaemia, evidence from NCRN CLL207: a phase II trial assessing alemtuzumab consolidation. Br J Haematol 2016; 176:573-582. [DOI: 10.1111/bjh.14342] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/13/2016] [Indexed: 12/21/2022]
Affiliation(s)
| | - Dena R. Howard
- Leeds Institute of Clinical Trials Research; University of Leeds; Leeds UK
| | | | - Andy C. Rawstron
- Department of Haematology; Leeds Teaching Hospitals NHS Trust; Leeds UK
- Hull York Medical School; University of York; York UK
| | | | | | | | | | - Donald Milligan
- Centre for Haematology and Stem Cell Transplantation; Heartlands Hospital; Birmingham UK
| | - Alexandra F. Smith
- Leeds Institute of Clinical Trials Research; University of Leeds; Leeds UK
| | - Walter Gregory
- Leeds Institute of Clinical Trials Research; University of Leeds; Leeds UK
| | - Peter Hillmen
- Department of Haematology; Leeds Teaching Hospitals NHS Trust; Leeds UK
| | | |
Collapse
|
26
|
Alemtuzumab Induction and Delayed Acute Rejection in Steroid-Free Simultaneous Pancreas-Kidney Transplant Recipients. Transplant Direct 2016; 3:e124. [PMID: 28349124 PMCID: PMC5361562 DOI: 10.1097/txd.0000000000000634] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/13/2016] [Indexed: 01/07/2023] Open
Abstract
Supplemental digital content is available in the text. Background The optimal immunosuppressive regimen in simultaneous pancreas-kidney transplant (SPKT) recipients that prevents acute rejection episodes (AREs) and allows optimal outcome remains elusive. Methods This cohort study assessed incidence and time to AREs in 73 consecutive SPKT recipients receiving alemtuzumab induction and steroid-free maintenance with tacrolimus and mycophenolate mofetil. A cohort with single high-dose antithymocyte globulin (ATG; n = 85) and triple therapy served as controls. In addition, we provided mechanistic insights in AREs after alemtuzumab depletion, including composition and alloreactivity of lymphocytes (flow cytometry and mixed lymphocyte reaction) plasma alemtuzumab levels (enzyme-linked immunosorbent assay), and maintenance drug exposure. Results Overall number of AREs at 3 years was significantly lower with alemtuzumab versus ATG induction (26.0% vs 43.5%; adjusted hazard ratio, 0.38; P = 0.029). Most AREs (94.6%) with ATG occurred within the first month, whereas 84.2% of AREs with alemtuzumab occurred beyond 3 months. Patients with and without an ARE in the steroid-free alemtuzumab group showed no differences in composition of lymphocytes, or in alemtuzumab levels. Of note, more than two thirds of these AREs were preceded by empiric tacrolimus and/or mycophenolate mofetil dose adjustments due to viral infections, leukopenia, or gastrointestinal symptoms. Conclusions Alemtuzumab induction resulted in a significant lower incidence of AREs. Empiric dose adjustments beyond 3 months in the absence of steroids carry a significant risk for subsequent rejection in SPKT recipients.
Collapse
|
27
|
Zhang L, Navaratna T, Thurber GM. A Helix-Stabilizing Linker Improves Subcutaneous Bioavailability of a Helical Peptide Independent of Linker Lipophilicity. Bioconjug Chem 2016; 27:1663-72. [PMID: 27327034 DOI: 10.1021/acs.bioconjchem.6b00209] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stabilized peptides address several limitations to peptide-based imaging agents and therapeutics such as poor stability and low affinity due to conformational flexibility. There is also active research in developing these compounds for intracellular drug targeting, and significant efforts have been invested to determine the effects of helix stabilization on intracellular delivery. However, much less is known about the impact on other pharmacokinetic parameters such as plasma clearance and bioavailability. We investigated the effect of different fluorescent helix-stabilizing linkers with varying lipophilicity on subcutaneous (sc) bioavailability using the glucagon-like peptide-1 (GLP-1) receptor ligand exendin as a model system. The stabilized peptides showed significantly higher protease resistance and increased bioavailability independent of linker hydrophilicity, and all subcutaneously delivered conjugates were able to successfully target the islets of Langerhans with high specificity. The lipophilic peptide variants had slower absorption and plasma clearance than their respective hydrophilic conjugates, and the absolute bioavailability was also lower likely due to the longer residence times in the skin. Their ease and efficiency make double-click helix stabilization chemistries a useful tool for increasing the bioavailability of peptide therapeutics, many of which suffer from rapid in vivo protease degradation. Helix stabilization using linkers of varying lipophilicity can further control sc absorption and clearance rates to customize plasma pharmacokinetics.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Tejas Navaratna
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Greg M Thurber
- Department of Chemical Engineering, and ‡Department of Biomedical Engineering University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
28
|
Pharmacokinetics and immunological outcomes of alemtuzumab-based treatment for steroid-refractory acute GvHD. Bone Marrow Transplant 2016; 51:1153-5. [PMID: 27042838 DOI: 10.1038/bmt.2016.83] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
29
|
Alemtuzumab and CHOP Chemotherapy for the Treatment of Aggressive Histology Peripheral T Cell Lymphomas: A Multi-Center Phase I Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2016; 16:18-28.e4. [DOI: 10.1016/j.clml.2015.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 10/01/2015] [Accepted: 11/12/2015] [Indexed: 02/02/2023]
|
30
|
|
31
|
Eskander RN, Tewari KS. Development of bevacizumab in advanced cervical cancer: pharmacodynamic modeling, survival impact and toxicology. Future Oncol 2015; 11:909-22. [PMID: 25760973 DOI: 10.2217/fon.14.276] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Historically, patients with metastatic, persistent or recurrent cervical cancer had limited therapeutic options. Despite several Phase II/III clinical trials, the combination of cisplatin and paclitaxel remained the most effective chemotherapeutic regimen. In 2014, publication of Gynecologic Oncology Group 240 represented the emergence of an alternate and effective therapeutic option. This prospective, randomized, Phase III clinical trial explored the impact of adding the antiangiogenic agent bevacizumab to two separate cytotoxic chemotherapy backbones. Importantly, the study met its primary end point, showing a survival advantage of approximately 4 months without detriment in quality of life. As such, a review of bevacizumab and its application in patients with advanced-stage cervical cancer is warranted.
Collapse
Affiliation(s)
- Ramez N Eskander
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, University of California, Irvine Medical Center, Orange, CA 92868, USA
| | | |
Collapse
|
32
|
Oude Munnink TH, Henstra MJ, Segerink LI, Movig KLL, Brummelhuis-Visser P. Therapeutic drug monitoring of monoclonal antibodies in inflammatory and malignant disease: Translating TNF-α experience to oncology. Clin Pharmacol Ther 2015; 99:419-31. [PMID: 26265133 DOI: 10.1002/cpt.211] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/21/2015] [Accepted: 08/07/2015] [Indexed: 12/22/2022]
Abstract
Lack of response to monoclonal antibodies (mAbs) has been associated with inadequate mAb serum concentrations. Therapeutic drug monitoring (TDM) of mAbs has the potential to guide to more effective dosing in individual patients. This review discusses the mechanisms responsible for interpatient variability of mAb pharmacokinetics, summarizes exposure-response data of mAbs used in inflammatory and malignant disease, presents current evidence of mAb-TDM in inflammatory disease, and provides hurdles and required future steps for further implementing mAb-TDM.
Collapse
Affiliation(s)
- T H Oude Munnink
- Department of Clinical Pharmacy, Medisch Spectrum Twente, Enschede, The Netherlands.,Department of Clinical Pharmacy, Hospital Group Twente, Almelo-Hengelo, The Netherlands
| | - M J Henstra
- Department of Clinical Pharmacy, Hospital Group Twente, Almelo-Hengelo, The Netherlands
| | - L I Segerink
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology and MIRA Institute for Biomedical Engineering and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - K L L Movig
- Department of Clinical Pharmacy, Medisch Spectrum Twente, Enschede, The Netherlands
| | - P Brummelhuis-Visser
- Department of Clinical Pharmacy, Hospital Group Twente, Almelo-Hengelo, The Netherlands
| |
Collapse
|
33
|
A Phase 2 Trial of Fludarabine Combined With Subcutaneous Alemtuzumab for the Treatment of Relapsed/Refractory B-Cell Chronic Lymphocytic Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2015; 15:694-8. [DOI: 10.1016/j.clml.2015.07.640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/01/2015] [Accepted: 07/28/2015] [Indexed: 11/19/2022]
|
34
|
Patel K, Parmar S, Shah S, Shore T, Gergis U, Mayer S, van Besien K. Comparison of Subcutaneous versus Intravenous Alemtuzumab for Graft-versus-Host Disease Prophylaxis with Fludarabine/Melphalan-Based Conditioning in Matched Unrelated Donor Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2015; 22:456-61. [PMID: 26524732 PMCID: PMC7128235 DOI: 10.1016/j.bbmt.2015.10.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/23/2015] [Indexed: 11/27/2022]
Abstract
The objective of this study was to compare infusion-related reactions and outcomes of using subcutaneous (subQ) alemtuzumab versus intravenous (i.v.) alemtuzumab as graft-versus-host disease (GVHD) prophylaxis for matched unrelated donor stem cell transplantations. Outcomes include incidence of cytomegalovirus (CMV)/Epstein-Barr (EBV) viremia, development of CMV disease or post-transplantation lymphoproliferative disorder, fatal infections, acute and chronic GVHD, time to engraftment, relapse rate, and survival. We conducted a retrospective study of all adult matched unrelated donor stem cell transplantations patients who received fludarabine/melphalan with subQ or i.v. alemtuzumab in combination with tacrolimus as part of their conditioning for unrelated donor transplantation at New York-Presbyterian/Weill Cornell Medical Center from January 1, 2012 to March 21, 2014. Alemtuzumab was administered at a total cumulative dose of 100 mg (divided over days −7 to −3). Forty-six patients received an unrelated donor stem cell transplantation with fludarabine/melphalan and either subQ (n = 26) or i.v. (n = 20) alemtuzumab in combination with tacrolimus. Within the evaluable population, 130 subQ and 100 i.v. alemtuzumab doses were administered. For the primary outcome, ≥grade 2 infusion-related reactions occurred in 11 (8%) versus 25 (25%) infusions in the subQ and i.v. cohorts, respectively (P = .001). Overall, 12 injections (9%) in the subQ arm versus 26 infusions (26%) in the i.v. arm experienced an infusion-related reaction of any grade (P = .001). There were no significant differences between the subQ and i.v. arms in rates of reactivation of CMV/EBV, development of CMV disease or post-transplantation lymphoproliferative disorder, fatal infections, acute and chronic GVHD, relapse, or survival. Subcutaneous administration of alemtuzumab for GVHD prophylaxis was associated with fewer infusion-related reactions compared with i.v. administration in the SCT setting. Incidences of acute and chronic GVHD were similar between both arms. There was also no difference in reactivation of CMV/EBV viremia, development of CMV disease or post-transplantation lymphoproliferative disorder, fatal infections, relapse, or survival. We conducted a retrospective comparison of intravenous versus subcutaneous administration of alemtuzumab for graft-versus-host disease prophylaxis in unrelated donor transplant recipients. We observed a significant reduction in grade ≥ 2 infusion-related reactions after subcutaneous injections. The incidences of acute and chronic graft-versus-host disease were similar. There was no difference in incidence of viral reactivation or fatal bacterial infections. There was no difference in 30-day mortality or 1-year overall survival between the 2 arms.
Collapse
Affiliation(s)
- Khilna Patel
- Department of Pharmacy, New York-Presbyterian Hospital, New York, New York.
| | - Sapna Parmar
- Department of Pharmacy, New York-Presbyterian Hospital, New York, New York
| | - Shreya Shah
- Department of Pharmacy, New York-Presbyterian Hospital, New York, New York
| | - Tsiporah Shore
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| | - Usama Gergis
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| | - Sebastian Mayer
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| | - Koen van Besien
- Department of Medicine, New York-Presbyterian Hospital, New York, New York
| |
Collapse
|
35
|
Damlaj M, Sulai NH, Oliveira JL, Ketterling RP, Hashmi S, Witzig T, Nowakowski G, Call TG, Shanafelt TD, Ding W, Hogan WJ, Litzow MR, Patnaik MM. Impact of Alemtuzumab Therapy and Route of Administration in T-Prolymphocytic Leukemia: A Single-Center Experience. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2015; 15:699-704. [PMID: 26422251 DOI: 10.1016/j.clml.2015.07.643] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/06/2015] [Accepted: 07/28/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We conducted a single-center retrospective analysis to determine the impact of the anti-CD52 monoclonal antibody alemtuzumab including route of administration compared to non-alemtuzumab-containing regimens in T-prolymphocytic leukemia (T-PLL). PATIENTS AND METHODS The study was a retrospective analysis of a consecutive cohort of adult patients diagnosed with T-PLL at Mayo Clinic Rochester from January 1, 1997, through September 30, 2014. RESULTS A total of 41 patients were diagnosed with T-PLL per the World Health Organization 2008 classification. The median age was 66 years, and 23 (56%) were male. After a median follow-up of 18 months (range, 0.4-66.1 months), 32 patients (78%) had died, with a median overall survival of 16.9 months. Approximately half the cohort was treated with alemtuzumab, almost exclusively after 2004. Median survival for patients receiving intravenous alemtuzumab-based therapy was 40.5 versus 10.3 months for all other therapies (P = .0004). A significant survival difference between intravenous versus subcutaneous alemtuzumab administration of 40.5 versus 13.7 months was noted (P = .0014). Only 4 (14%) of 28 patients aged < 70 years underwent hematopoietic stem cell transplantation, with a median survival after transplantation of 4 months. CONCLUSION In this large series of T-PLL patients treated at a single tertiary-care center, we confirmed the prior observation of the superiority of intravenous alemtuzumab over other therapies. Hematopoietic stem cell transplantation was feasible in a minority of potentially eligible patients. Early transplant referral should be considered for all eligible patients.
Collapse
Affiliation(s)
- Moussab Damlaj
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN.
| | - Nanna H Sulai
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Jennifer L Oliveira
- Division of Hematopathology, Department of Laboratory Medicine, Mayo Clinic, Rochester, MN
| | - Rhett P Ketterling
- Division of Hematopathology, Department of Laboratory Medicine, Mayo Clinic, Rochester, MN
| | - Shahrukh Hashmi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Thomas Witzig
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | | | - Timothy G Call
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Tait D Shanafelt
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Wei Ding
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - William J Hogan
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Mark R Litzow
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Mrinal M Patnaik
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
36
|
Deppisch N, Ruf P, Eissler N, Neff F, Buhmann R, Lindhofer H, Mocikat R. Efficacy and Tolerability of a GD2-Directed Trifunctional Bispecific Antibody in a Preclinical Model: Subcutaneous Administration Is Superior to Intravenous Delivery. Mol Cancer Ther 2015; 14:1877-83. [PMID: 26063765 DOI: 10.1158/1535-7163.mct-15-0156] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 06/03/2015] [Indexed: 11/16/2022]
Abstract
Trifunctional bispecific antibodies (trAb) are novel anticancer drugs that recruit and activate different types of immune effector cells at the targeted tumor. Thus, tumor cells are effectively eliminated and a long-lasting tumor-specific T-cell memory is induced. The trAb Ektomab is directed against human CD3 on T cells and the tumor-associated ganglioside GD2, which is an attractive target for immunotherapy of melanoma in humans. To optimize clinical applicability, we studied different application routes with respect to therapeutic efficacy and tolerability by using the surrogate trAb Surek (anti-GD2 × anti-murine CD3) and a murine melanoma engineered to express GD2. We show that subcutaneous injection of the trAb is superior to the intravenous delivery pathway, which is the standard application route for therapeutic antibodies. Despite lower plasma levels after subcutaneous administration, the same tumor-protective potential was observed in vivo compared with intravenous administration of Surek. However, subcutaneously delivered Surek showed better tolerability. This could be explained by a continuous release of the antibody leading to constant plasma levels and a delayed induction of proinflammatory cytokines. Importantly, the induction of counter-regulatory mechanisms was reduced after subcutaneous application. These findings are relevant for the clinical application of trifunctional bispecific antibodies and, possibly, also other immunoglobulin constructs. Mol Cancer Ther; 14(8); 1877-83. ©2015 AACR.
Collapse
Affiliation(s)
- Nina Deppisch
- Helmholtz-Zentrum München, Institut für Molekulare Immunologie, Munich, Germany
| | - Peter Ruf
- Trion Research GmbH, Martinsried, Germany
| | - Nina Eissler
- Helmholtz-Zentrum München, Institut für Molekulare Immunologie, Munich, Germany
| | - Frauke Neff
- Helmholtz-Zentrum München, Institut für Pathologie, Munich, Germany
| | - Raymund Buhmann
- Ludwig-Maximilians-Universität München, Klinikum Großhadern, Medizinische Klinik III und Abteilung für Transfusionsmedizin, Munich, Germany
| | | | - Ralph Mocikat
- Helmholtz-Zentrum München, Institut für Molekulare Immunologie, Munich, Germany.
| |
Collapse
|
37
|
Marsh RA, Rao MB, Gefen A, Bellman D, Mehta PA, Khandelwal P, Chandra S, Jodele S, Myers KC, Grimley M, Dandoy C, El-Bietar J, Kumar AR, Leemhuis T, Zhang K, Bleesing JJ, Jordan MB, Filipovich AH, Davies SM. Experience with Alemtuzumab, Fludarabine, and Melphalan Reduced-Intensity Conditioning Hematopoietic Cell Transplantation in Patients with Nonmalignant Diseases Reveals Good Outcomes and That the Risk of Mixed Chimerism Depends on Underlying Disease, Stem Cell Source, and Alemtuzumab Regimen. Biol Blood Marrow Transplant 2015; 21:1460-70. [PMID: 25865646 DOI: 10.1016/j.bbmt.2015.04.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/02/2015] [Indexed: 10/23/2022]
Abstract
Alemtuzumab, fludarabine, and melphalan reduced-intensity conditioning (RIC) regimens are increasingly used for the hematopoietic cell transplantation (HCT) of pediatric and young adult patients with nonmalignant diseases. Early experience suggests that these regimens are associated with good survival but a high incidence of mixed chimerism, which we have previously shown to be influenced by the alemtuzumab schedule. We hypothesized that the underlying diagnosis and donor graft source would also affect the development of mixed chimerism and that the majority of patients would survive RIC HCT without graft loss. To examine this, we conducted a retrospective study of 206 patients with metabolic diseases, non-Fanconi anemia marrow failure disorders, and primary immune deficiencies who underwent 210 consecutive RIC HCT procedures at Cincinnati Children's Hospital. Ninety-seven percent of the patients engrafted. Mixed donor and recipient chimerism developed in 46% of patients. Patients with marrow failure had a low risk of mixed chimerism (hazard ratio [HR], .208; 95% confidence interval [CI], .061 to .709; P = .012). The risk of mixed chimerism was high in patients who received a cord blood graft (HR, 3.122; 95% CI, 1.236 to 7.888; P = .016). As expected, patients who received a proximal or higher dose per kilogram of alemtuzumab schedule also experienced higher rates of mixed chimerism (all HR > 2, all P < .05). At the time of last follow-up (median, 654 days; range, 13 to 3337), over 75% of patients had greater than 90% whole blood donor chimerism. A second transplantation was performed in 5% of patients. Three-year survival without retransplantation was 84% (95% CI, 71% to 98%) for patients who underwent transplantation with an HLA-matched sibling donor. Survival without retransplantation was negatively affected by lack of a matched related donor, increasing age, and development of grades III and IV acute graft-versus-host disease. We conclude that alemtuzumab, fludarabine, and melphalan RIC HCT offers good results for many patients and that the risk of developing mixed chimerism is influenced by underlying diagnosis, graft source, and alemtuzumab dosing.
Collapse
Affiliation(s)
- Rebecca A Marsh
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | - Marepalli B Rao
- Division of Epidemiology and Biostatistics, Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Aharon Gefen
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Denise Bellman
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Parinda A Mehta
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Pooja Khandelwal
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sharat Chandra
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kasiani C Myers
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael Grimley
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christopher Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Javier El-Bietar
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ashish R Kumar
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tom Leemhuis
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kejian Zhang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jack J Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael B Jordan
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alexandra H Filipovich
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
38
|
AlDeghaither D, Smaglo BG, Weiner LM. Beyond peptides and mAbs--current status and future perspectives for biotherapeutics with novel constructs. J Clin Pharmacol 2015; 55 Suppl 3:S4-20. [PMID: 25707963 PMCID: PMC4340091 DOI: 10.1002/jcph.407] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/29/2014] [Indexed: 12/26/2022]
Abstract
Biotherapeutics are attractive anti-cancer agents due to their high specificity and limited toxicity compared to conventional small molecules. Antibodies are widely used in cancer therapy, either directly or conjugated to a cytotoxic payload. Peptide therapies, though not as prevalent, have been utilized in hormonal therapy and imaging. The limitations associated with unmodified forms of both types of biotherapeutics have led to the design and development of novel structures, which incorporate key features and structures that have improved the molecules' abilities to bind to tumor targets, avoid degradation, and exhibit favorable pharmacokinetics. In this review, we highlight the current status of monoclonal antibodies and peptides, and provide a perspective on the future of biotherapeutics using novel constructs.
Collapse
Affiliation(s)
- Dalal AlDeghaither
- Georgetown Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington DC 20057
| | - Brandon G Smaglo
- Medstar Georgetown University Hospital, Department of Medicine, Division of Hematology/Oncology, 3800 Reservoir Road NW, Washington DC 20007
| | - Louis M. Weiner
- Georgetown Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University Medical Center, 3970 Reservoir Road NW, Washington DC 20057
| |
Collapse
|
39
|
Richter WF, Jacobsen B. Subcutaneous absorption of biotherapeutics: knowns and unknowns. Drug Metab Dispos 2014; 42:1881-9. [PMID: 25100673 DOI: 10.1124/dmd.114.059238] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Subcutaneous administration of biotherapeutics offers several potential advantages compared with intravenous administration. Many biotherapeutics, both marketed or in development, are administered via the subcutaneous route. This minireview provides an overview of the presystemic absorption processes following subcutaneous administration, the resulting pharmacokinetics after subcutaneous administration, and provides recent case examples of the development of subcutaneous administered drugs with a focus on monoclonal antibodies. Subcutaneous absorption of biotherapeutics is relatively slow and mostly incomplete. Knowledge of the subcutaneous tissue is important to understand the absorption kinetics after subcutaneous administration. Transport in the subcutis to the absorbing blood or lymph capillaries appears to be a major contributor to the slow subcutaneous absorption. Larger proteins (>20 kDa) are mostly absorbed via the lymphatic system, although potential species differences are not fully understood yet. Also, the presystemic catabolism leading to incomplete bioavailability is little understood, both the involved enzymes and its translation across species. For IgGs, binding to the neonatal Fc receptor is important to obtain a high bioavailability. Overall, several aspects of subcutaneous absorption are still poorly understood, which hampers, e.g., translation across species. Further research in this area is warranted.
Collapse
Affiliation(s)
- Wolfgang F Richter
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Björn Jacobsen
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
40
|
Outcomes of hematopoietic cell transplantation in adult patients with acquired aplastic anemia using intermediate-dose alemtuzumab-based conditioning. Biol Blood Marrow Transplant 2014; 20:1722-8. [PMID: 25017761 DOI: 10.1016/j.bbmt.2014.06.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/25/2014] [Indexed: 11/21/2022]
Abstract
Graft-versus-host disease (GVHD) has no therapeutic benefit after hematopoietic cell transplantation (HCT) for patients with acquired aplastic anemia (AA), and its prevention is highly desirable. We designed a conditioning regimen using an intermediate dose of alemtuzumab (50 to 60 mg) and describe our institutional experience of 41 patients who underwent HCT for AA. The median age at HCT was 37 years (range, 17 to 59). The conditioning regimen was high-dose cyclophosphamide (n = 9) or fludarabine based (n = 32). Additional GVHD prophylaxis was with cyclosporine. With a median follow-up of 3.6 years, overall survival at 3 years was 85%. Survival in patients <40 years and ≥40 years was 96% and 67%, respectively (P = .04). Graft failure occurred in 4 (10%) patients; 2 primary and 2 secondary. The cumulative incidences of acute (grades 1 to 2) and chronic GVHD were 27% and 15%, respectively. No patients developed grade 3 to 4 acute GVHD or severe chronic GVHD. The following viral complications were frequent: cytomegalovirus reactivation (79%), herpes simplex (18%), varicella zoster (25%), and BK virus hemorrhagic cystitis (8%). The majority of patients had no significant long-term health issues. This intermediate-dose alemtuzumab-based conditioning regimen results in excellent survival with a favorable impact on GVHD and long-term health outcomes, but close monitoring for viral complications is important.
Collapse
|
41
|
Jackisch C, Müller V, Maintz C, Hell S, Ataseven B. Subcutaneous Administration of Monoclonal Antibodies in Oncology. Geburtshilfe Frauenheilkd 2014; 74:343-349. [PMID: 25076790 DOI: 10.1055/s-0034-1368173] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/20/2014] [Accepted: 02/02/2014] [Indexed: 12/29/2022] Open
Abstract
Treatment with monoclonal antibodies (mabs) has become an established component of oncological therapy. The monoclonal antibodies available for this purpose are mainly administered intravenously in individually adapted doses according to body weight over longer treatment times. For other chronic diseases such as, for example, diabetes mellitus, the subcutaneous administration of drugs is an established therapy option. For the subcutaneous administration of larger volumes as needed for mab solutions the extracellular matrix of the subcutaneous tissue represents a problem. The co-formulation with recombinant human hyaluronidase makes the relatively pain-free administration of larger fluid volumes and thus the subcutaneous administration of monoclonal antibodies possible, as illustrated by the development of a subcutaneous formulation of trastuzumab. This constitutes a less invasive, time-optimised and flexible form of administration for patients with HER2-positive breast cancer that, with its fixed dosing possibilities, contributes to therapeutic safety. The example of trastuzumab shows that the subcutaneous administration of monoclonal antibodies can simplify oncological long-term therapy not only for the patients but also for the medical personnel.
Collapse
Affiliation(s)
- C Jackisch
- Obstetrics and Gynecology, Sanaklinikum Offenbach, Offenbach
| | - V Müller
- Klinik und Poliklinik für Gynäkologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg
| | - C Maintz
- Hämatologisch-Onkologische Praxis, Würselen
| | - S Hell
- Medizinisch-wissenschaftliche Beratung, Speyer
| | - B Ataseven
- Klinik für Gynäkologie & Gynäkologische Onkologie, Kliniken Essen-Mitte, Essen
| |
Collapse
|
42
|
Abstract
Alemtuzumab (formerly known as Campath-1H) has recently been approved by the European Medicines Agency for highly-active, relapsing-remitting multiple sclerosis (MS). The molecule targets the CD52 surface glycoprotein on certain T cells and B cells and is thought to exert its effect in MS through a “resetting” of the lymphocyte population. Approval was granted on the strength of two pivotal studies, Comparison of Alemtuzumab and Rebif® Efficacy in Multiple Sclerosis (CARE-MS)-1 in the first-line setting and CARE-MS-2 in patients who had failed first-line therapy. In both studies, alemtuzumab significantly reduced the relapse rate compared to the comparator, interferon beta-1a (44 μg) given subcutaneously three-times per week (Rebif®). In the first-line study, alemtuzumab was also found to significantly reduce the number of patients with sustained progression compared to interferon beta-1a therapy. Autoimmune disorders represent the major side effect of alemtuzumab therapy although they can be managed by careful monitoring and early treatment. Overall, alemtuzumab is likely to be a valuable addition to the neurologist’s armamentarium for the treatment of relapsing-remitting MS.
Collapse
Affiliation(s)
- Oscar Fernandez
- Institute of Clinical Neuroscience, Neurology Department, Hospital Regional Universitario Carlos Haya, FIMABIS, Malaga, Spain
| |
Collapse
|
43
|
Hale GA. Autologous hematopoietic stem cell transplantation for pediatric solid tumors. Expert Rev Anticancer Ther 2014; 5:835-46. [PMID: 16221053 DOI: 10.1586/14737140.5.5.835] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
While advances in the treatment of pediatric cancers have increased cure rates, children with metastatic or recurrent solid tumors have a dismal prognosis despite initial transient responses to therapy. Autologous hematopoietic stem cell transplantation takes advantage of the steep dose-response relationship observed with many chemotherapeutic agents. While clearly demonstrated to improve outcomes in patients with metastatic neuroblastoma, autologous hematopoietic stem cell transplantation is also frequently used to treat patients with other high-risk diseases such as Ewing sarcoma, osteosarcoma, rhabdomyosarcoma, Wilms' tumor, retinoblastoma, germ cell tumors, lymphomas and brain tumors. Most published experience consists of retrospective, single-arm studies; randomized clinical trials are lacking, due in part to the rarity of pediatric cancers treatable by autologous hematopoietic stem cell transplantation. These published literature demonstrate that autologous hematopoietic stem cell transplantation results in most cases in equivalent or superior outcomes when compared with conventional therapies. However, patient heterogeneity, patient selection, graft characteristics and processing and the varied conditioning regimens are additional factors to consider. Since the inception of autologous hematopoietic stem cell transplantation, regimen-related toxicity has markedly decreased and the vast majority of treatment failures are now due to disease recurrence. Prospective clinical trials are needed to identify specific high-risk patient populations, with randomization (when possible) to compare outcomes of patients undergoing autologous hematopoietic stem cell transplantation with those receiving standard therapy. In addition, investigators need to better define the role of autologous hematopoietic stem cell transplantation in these solid tumors, particularly in combination with other therapeutic modalities such as immunotherapy and novel cell processing methodologies.
Collapse
Affiliation(s)
- Gregory A Hale
- Department of Hematology/Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
44
|
|
45
|
Costelloe L, Jones J, Coles A. Secondary autoimmune diseases following alemtuzumab therapy for multiple sclerosis. Expert Rev Neurother 2014; 12:335-41. [DOI: 10.1586/ern.12.5] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Novitzky N, Davison G, Abdulla R, Mowla S. Definition of the Variables Affecting Efficacy of Immunodepletion Ex Vivo of Peripheral Blood Progenitor Cell Grafts by Alemtuzumab (Campath in the Bag). Biol Blood Marrow Transplant 2013; 19:1753-9. [DOI: 10.1016/j.bbmt.2013.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 10/01/2013] [Indexed: 11/25/2022]
|
47
|
Reiff A. A review of Campath in autoimmune disease: Biologic therapy in the gray zone between immunosuppression and immunoablation. Hematology 2013; 10:79-93. [PMID: 16019453 DOI: 10.1080/10245330400026139] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Campath, the subject of this review, is an example of a broadly targeted biologic agent, approved for patients with B-CLL, which may combine immunosuppressive as well as immunoablative properties. For many years Campath has been in clinical use as an immunosuppressive agent for various autoimmune diseases and as part of the preparative regimes for allogeneic HSCT, successfully preventing graft-versus-host-disease (GVHD). This review summarizes the experience of 24 studies including a total of 323 patients treated with Campath for various autoimmune diseases such as arthritis, MS, vasculitis, autoimmune cytopenias and others. The results demonstrate that Campath is fairly safe and 75% of the patients experienced clinical improvement and 15% of the patients were reported in clinical remission even though improvements were often transient.While other biologic drugs may have to narrow targets, Campath, is able to bridge the gap between immunosuppression and immunoablation and may offer an alternative to human stem cell transplantation avoiding the risks of chemotherapy and radiation.
Collapse
Affiliation(s)
- Andreas Reiff
- Division of Rheumatology, Department of Pediatrics, Keck School of Medicine, University of Southern California, Childrens Hospital, Los Angeles 90027, USA.
| |
Collapse
|
48
|
Mellstedt H. Anti-neoplastic biosimilars—the same rules as for cytotoxic generics cannot be applied. Ann Oncol 2013; 24 Suppl 5:v23-8. [DOI: 10.1093/annonc/mdt325] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
49
|
Abstract
Alemtuzumab (Campath®, MabCampath®, Genzyme) is an IgG1k anti-CD52 humanized monoclonal antibody (mAb) that was first licensed in March 2001 by FDA. EMEA granted its approval in July 2001 and Health Canada in November 2005. The initial indication was limited to B-CLL previously treated and resistant to alkylating agents. Starting from 2007, alemtuzumab was approved also as first-line therapy of B-CLL. So far, it has been experienced in over 60 countries.
Collapse
|
50
|
|