Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Blood ; 143(11): 1049-1054, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38052031

ABSTRACT

ABSTRACT: We show that red cell exchange (RCE) treats hyperleukocytosis in acute leukemia. RCE provided similar leukoreduction to standard therapeutic leukoreduction and could be superior in patients with severe anemia or monocytic leukemias or when requiring rapid treatment.


Subject(s)
Leukemia, Monocytic, Acute , Leukemia, Myeloid, Acute , Leukostasis , Adult , Humans , Leukostasis/therapy , Leukemia, Myeloid, Acute/therapy , Leukemia, Monocytic, Acute/therapy , Acute Disease , Leukapheresis , Leukocytosis/therapy
2.
J Clin Apher ; 38(2): 77-278, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37017433

ABSTRACT

The American Society for Apheresis (ASFA) Journal of Clinical Apheresis (JCA) Special Issue Writing Committee is charged with reviewing, updating, and categorizing indications for the evidence-based use of therapeutic apheresis (TA) in human disease. In the Ninth Edition, the JCA Special Issue Writing Committee has incorporated systematic review and evidence-based approaches in the grading of evidence and categorization of apheresis indications to make recommendations on the use of apheresis in a wide variety of diseases and conditions. This edition has largely maintained the general layout and concept of a fact sheet introduced in the Fourth Edition (2007). Each fact sheet succinctly summarizes the evidence for the use of TA in a specific disease or medical condition. The Ninth Edition of the JCA Special Issue comprises 91 fact sheets and 166 graded and categorized indications. This includes seven new fact sheets, nine new indications on existing fact sheets, and eight changes in the category for existing indications. The Ninth Edition of the JCA Special Issue seeks to continue to serve as a key resource that guides the utilization of TA in the treatment of human disease.


Subject(s)
Blood Component Removal , Evidence-Based Medicine , Humans , United States , Writing
3.
Cytotherapy ; 24(9): 916-922, 2022 09.
Article in English | MEDLINE | ID: mdl-35398001

ABSTRACT

BACKGROUND AIMS: This white paper was developed to provide leukapheresis guidance for the collection of mononuclear cells from adult and pediatric patients who are destined for immune effector cell (IEC) therapies for commercial and research applications. Currently, there is considerable variability in leukapheresis processes and limited published information regarding best practices relevant to new cellular therapies, especially IECs. Herein the authors address critical leukapheresis questions in five domains to help guide consistent collection processes and ensure high-quality products. The first four domains are onboarding, pre-collection, collection and post-collection, with protocol feasibility, preparation, care and follow-up of the patient/donor at each step, respectively, and technical considerations during collection. The fifth domain of quality assurance focuses on ensuring product potency, purity, safety and auditing. METHODS: The American Society for Apheresis (ASFA) Clinical Applications Committee (IEC Therapy Subcommittee) was charged by the society's board of directors with working collaboratively with other ASFA committees and organizations, including the Foundation for the Accreditation of Cellular Therapy, Association for the Advancement of Blood and Biotherapies, American Society for Transplantation and Cellular Therapy, National Marrow Donor Program and International Society for Cell & Gene Therapy, to develop guidelines regarding leukapheresis collection of cells destined for the manufacture of IEC therapies. After a review of the literature and discussion with members of the involved committees and various institutions, a draft guidance was created and circulated for comment and revision. RESULTS: Critical aspects of apheresis that could affect the quality and quantity of the leukapheresis product were identified. These areas were then discussed and reviewed. After consensus, the best practice guidelines were proposed and accepted. CONCLUSIONS: In the current era of rapid growth of IEC therapies, it is important to address critical leukapheresis steps to provide high-quality products and more consistent practices and to eliminate redundant efforts.


Subject(s)
Blood Component Removal , Adult , Blood Component Removal/methods , Cell- and Tissue-Based Therapy , Child , Consensus , Humans , Leukapheresis/methods , Tissue Donors , United States
4.
J Clin Apher ; 37(3): 223-236, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35085413

ABSTRACT

BACKGROUND: Chimeric antigen receptor T (CAR-T) cell successes have encouraged continued clinical study. Apheresis collection of starting material for CAR-T cell therapy product manufacturing is critical but described approaches suggest variability and clinical guidelines are currently lacking. The goal of this study was to gather and assess variability in apheresis collection descriptions in publicly available CAR T-cell therapy clinical trials. STUDY DESIGN: We searched clinicaltrials.gov (a publicly available clinical trial database) for "chimeric antigen receptor T cells" on July 01, 2020 and studies accessed July 30, 2020-August 15, 2020. Data collected included date posted, study characteristics, apheresis mentions (number, location, and context), laboratory parameters and transfusion allowances. Apheresis context was analyzed using a qualitative inductive approach of grounded theory method with open coding. Text was classified into 37 context codes, grouped into 12 categories, and then consolidated into patient, procedure, product, and miscellaneous themes. RESULTS: Apheresis was mentioned 1044 times in 322 (51.9%) of 621 total studies. Laboratory parameters mentioned included white blood cells (100 studies), absolute neutrophil count (220 studies), absolute lymphocyte count (102 studies), CD3+ cell (38 studies), hemoglobin (233 studies, 54 studies specified transfusion allowance), and platelet (269 studies, 48 studies specified transfusion allowance). CONCLUSIONS: Apheresis collection of CAR-T cell products is not well-defined in clinical study descriptions and the context is inconsistent. Laboratory parameters useful for apheresis collection are variably present and do not consistently align with current practices. Further exploration, and clinical guideline development will encourage alignment of apheresis collections for CAR-T cell products.


Subject(s)
Blood Component Removal , Receptors, Chimeric Antigen , Blood Component Removal/methods , Humans , Immunotherapy, Adoptive , Lymphocyte Count , T-Lymphocytes
5.
J Clin Invest ; 131(24)2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34788233

ABSTRACT

BackgroundAntibody-based strategies for COVID-19 have shown promise in prevention and treatment of early disease. COVID-19 convalescent plasma (CCP) has been widely used but results from randomized trials supporting its benefit in hospitalized patients with pneumonia are limited. Here, we assess the efficacy of CCP in severely ill, hospitalized adults with COVID-19 pneumonia.MethodsWe performed a randomized control trial (PennCCP2), with 80 adults hospitalized with COVID-19 pneumonia, comparing up to 2 units of locally sourced CCP plus standard care versus standard care alone. The primary efficacy endpoint was comparison of a clinical severity score. Key secondary outcomes include 14- and 28-day mortality, 14- and 28-day maximum 8-point WHO ordinal score (WHO8) score, duration of supplemental oxygenation or mechanical ventilation, respiratory SARS-CoV-2 RNA, and anti-SARS-CoV-2 antibodies.ResultsEighty hospitalized adults with confirmed COVID-19 pneumonia were enrolled at median day 6 of symptoms and day 1 of hospitalization; 60% were anti-SARS-CoV-2 antibody seronegative. Participants had a median of 3 comorbidities, including risk factors for severe COVID-19 and immunosuppression. CCP treatment was safe and conferred significant benefit by clinical severity score (median [MED] and interquartile range [IQR] 10 [5.5-30] vs. 7 [2.75-12.25], P = 0.037) and 28-day mortality (n = 10, 26% vs. n = 2, 5%; P = 0.013). All other prespecified outcome measures showed weak evidence toward benefit of CCP.ConclusionTwo units of locally sourced CCP administered early in hospitalization to majority seronegative participants conferred a significant benefit in clinical severity score and 28-day mortality. Results suggest CCP may benefit select populations, especially those with comorbidities who are treated early.Trial RegistrationClinicalTrials.gov NCT04397757.FundingUniversity of Pennsylvania.


Subject(s)
COVID-19/therapy , Pneumonia, Viral/therapy , SARS-CoV-2 , Adult , Aged , Antibodies, Viral , Female , Hospitalization , Humans , Immune Tolerance , Immunization, Passive/methods , Immunosuppression Therapy , Incidence , Male , Middle Aged , Oxygen/therapeutic use , RNA, Viral , Respiration, Artificial , Risk Factors , Treatment Outcome , COVID-19 Serotherapy
6.
J Clin Apher ; 35(5): 493-499, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32770558

ABSTRACT

Since 1986, the American Society for Apheresis (ASFA) has published practice guidelines on the use of therapeutic apheresis in the Journal of Clinical Apheresis (JCA) Special Issue. Since 2007, updated guidelines have been published every 3 years to reflect current evidence based apheresis practice with the most recent edition (8th) published in 2019. With each edition, the guidelines are reviewed and updated based on any newly published literature since the last review. The PEXIVAS study, an international, randomized controlled trial comparing therapeutic plasma exchange (TPE) vs no TPE and standard vs reduced dose steroid regimen on the primary composite outcome of end stage renal disease or death in patients with ANCA-associated vasculitis (AAV), was published in February 2020. This study represents the largest study on the role of therapeutic apheresis in AAV published to date and prompted the JCA Special Issue Writing Committee to reassess the current AAV fact sheet for updates based on this newly available evidence. This interim fact sheet summarizes current ASFA recommendations for the evidence-based use of therapeutic apheresis in AAV and supersedes the recommendations published in the 2019 guidelines.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/therapy , Blood Component Removal/methods , Practice Guidelines as Topic , Humans , Plasma Exchange , Societies, Medical
8.
J Clin Apher ; 34(3): 171-354, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31180581

ABSTRACT

The American Society for Apheresis (ASFA) Journal of Clinical Apheresis (JCA) Special Issue Writing Committee is charged with reviewing, updating and categorizing indications for the evidence-based use of therapeutic apheresis (TA) in human disease. Since the 2007 JCA Special Issue (Fourth Edition), the committee has incorporated systematic review and evidence-based approaches in the grading and categorization of apheresis indications. This Eighth Edition of the JCA Special Issue continues to maintain this methodology and rigor in order to make recommendations on the use of apheresis in a wide variety of diseases/conditions. The JCA Eighth Edition, like its predecessor, continues to apply the category and grading system definitions in fact sheets. The general layout and concept of a fact sheet that was introduced in the Fourth Edition, has largely been maintained in this edition. Each fact sheet succinctly summarizes the evidence for the use of TA in a specific disease entity or medical condition. The Eighth Edition comprises 84 fact sheets for relevant diseases and medical conditions, with 157 graded and categorized indications and/or TA modalities. The Eighth Edition of the JCA Special Issue seeks to continue to serve as a key resource that guides the utilization of TA in the treatment of human disease.


Subject(s)
Blood Component Removal/methods , Evidence-Based Medicine/standards , Humans , Therapeutics/methods , United States , Writing
9.
J Clin Apher ; 31(3): 149-62, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27322218

ABSTRACT

The American Society for Apheresis (ASFA) Journal of Clinical Apheresis (JCA) Special Issue Writing Committee is charged with reviewing, updating, and categorizing indications for the evidence-based use of therapeutic apheresis in human disease. Since the 2007 JCA Special Issue (Fourth Edition), the Committee has incorporated systematic review and evidence-based approaches in the grading and categorization of apheresis indications. This Seventh Edition of the JCA Special Issue continues to maintain this methodology and rigor to make recommendations on the use of apheresis in a wide variety of diseases/conditions. The JCA Seventh Edition, like its predecessor, has consistently applied the category and grading system definitions in the fact sheets. The general layout and concept of a fact sheet that was used since the fourth edition has largely been maintained in this edition. Each fact sheet succinctly summarizes the evidence for the use of therapeutic apheresis in a specific disease entity. The Seventh Edition discusses 87 fact sheets (14 new fact sheets since the Sixth Edition) for therapeutic apheresis diseases and medical conditions, with 179 indications, which are separately graded and categorized within the listed fact sheets. Several diseases that are Category IV which have been described in detail in previous editions and do not have significant new evidence since the last publication are summarized in a separate table. The Seventh Edition of the JCA Special Issue serves as a key resource that guides the utilization of therapeutic apheresis in the treatment of human disease. J. Clin. Apheresis 31:149-162, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Blood Component Removal/methods , Evidence-Based Medicine/standards , Practice Guidelines as Topic , Humans , Societies, Medical
10.
Am J Hematol ; 91(5): 453-60, 2016 May.
Article in English | MEDLINE | ID: mdl-26858124

ABSTRACT

Limited cell numbers in umbilical cord blood (UCB) grafts present a major impediment to favorable outcomes in adult transplantation, largely related to delayed or failed engraftment. The advent of UCB transplantation (UCBT) using two grafts successfully circumvents this obstacle, despite the engraftment of only one unit. Preclinical models suggested that the addition of UCB T cells at the time of transplant can enhance engraftment. We tested whether ex vivo activation by CD3/CD28 costimulation and expansion of T cells from a single UCB graft would be safe and feasible in adults with advanced hematologic malignancies, with an overall objective of optimizing engraftment in single unit UCBT. In this phase 1 study, recipients of single UCB units were eligible if the unit was stored in two adequate fractions. Dose limiting toxicity was defined as grade 3 or grade 4 GVHD within 90 days of UCBT. Four patients underwent UCBT; all were treated at the first dose level (10(5) cells/kg). At the 10(5) cells/kg dose level two subjects experienced grade 3 intestinal GVHD, thus meeting stopping criteria. For three subjects, neutrophil engraftment was early (12, 17, and 20 days), while one subject experienced primary graft failure. We observed early donor T cell trafficking and found that expanded T cells produced supraphysiologic levels of cytokines relevant to engraftment and to lymphoid differentiation and function. Taken together, these preliminary data suggest rapid engraftment in recipients of a single UCBT combined with relatively low doses of activated T cells, though potentially complicated by severe GVHD.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Lymphocyte Transfusion , T-Lymphocyte Subsets/transplantation , Adult , Anemia, Refractory, with Excess of Blasts/therapy , B-Cell Activating Factor/biosynthesis , Blood Preservation , CD28 Antigens/immunology , CD3 Complex/immunology , Cells, Cultured/transplantation , Cord Blood Stem Cell Transplantation/adverse effects , Cord Blood Stem Cell Transplantation/mortality , Cryopreservation , Cytokines/analysis , Dose-Response Relationship, Immunologic , Feasibility Studies , Female , Graft Survival , Graft vs Host Disease/etiology , Histocompatibility , Humans , Infant, Newborn , Leukemia, Myeloid, Acute/therapy , Lymphocyte Transfusion/adverse effects , Male , Maximum Tolerated Dose , Membrane Proteins/biosynthesis , Middle Aged , Neutrophils/transplantation , T-Cell Antigen Receptor Specificity , Transplantation Conditioning/methods , Treatment Outcome
11.
J Clin Apher ; 31(5): 448-53, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26575088

ABSTRACT

The thrombotic microangiopathy (TMA) Registry Network of North America (TRNA) is a collaborative network organized for the purpose of developing a multi-institutional registry and network to conduct clinical studies in a rare patient population. The TRNA was founded in 2013 by four academic medical centers (Columbia University Medical Center, Duke University Medical Center, University of Alabama at Birmingham, and University of Pennsylvania) to develop a national and demographically diverse dataset of patients with TMA. A clinical database was developed by network members using REDCap (Research Electronic Data Capture), a web-based database developed for clinical research. To facilitate rapid Institutional Review Board (IRB) approval at multiple sites, the TRNA utilized IRBshare, a streamlined IRB process to allow patient recruitment and enrollment into the TMA registry. This article reviews the process used to establish the TRNA network and discusses the significance of the first multi-institutional clinical apheresis network developed in the United States. J. Clin. Apheresis 31:448-453, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Blood Component Removal/methods , Registries , Thrombotic Microangiopathies/therapy , Academic Medical Centers , Databases, Factual , Humans , North America , United States
12.
Clin Cancer Res ; 21(8): 1835-42, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25632047

ABSTRACT

PURPOSE: Lenalidomide, an immunomodulatory agent that enhances antibody-dependent cell-mediated cytotoxicity, has the potential to synergize with rituximab, an anti-CD20 mAb. We hypothesized that the addition of lenalidomide to rituximab would improve clinical outcomes in patients with B-cell lymphomas who were previously rituximab resistant, defined as no response to or progression of lymphoma within 6 months of rituximab-based therapy. EXPERIMENTAL DESIGN: We conducted a single-center, phase II trial in patients with indolent B-cell or mantle cell lymphomas who were previously rituximab resistant. Patients received 10 mg lenalidomide daily for 8 weeks, and then received four weekly doses of 375 mg/m(2) rituximab; lenalidomide continued during and after rituximab. Response to therapy was assessed after 8 weeks of lenalidomide and 12 weeks after first dose of rituximab. The primary endpoint was overall response rate (ORR) after lenalidomide and rituximab. RESULTS: Fifty patients were enrolled and 43 patients completed both response assessments. ORR after 8 weeks of lenalidomide was 30.2%; 12 weeks after the addition of rituximab to lenalidomide, ORR increased to 62.8% (N = 43). For all patients (N = 50), median progression-free survival (PFS) is 22.2 months (median follow-up, 39.2 months). PFS after lenalidomide-rituximab was significantly longer than the PFS for the antecedent regimen used to define rituximab resistance (22.2 vs. 9.13 months, P = 0.0004). CONCLUSIONS: This trial is the first to show that the combination of lenalidomide and rituximab overcomes prior rituximab resistance in patients with indolent B-cell and mantle cell lymphomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Resistance, Neoplasm , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/pathology , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Female , Follow-Up Studies , Genotype , Humans , Lenalidomide , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/mortality , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Neoplasm Grading , Receptors, IgG/genetics , Retreatment , Rituximab/administration & dosage , T-Lymphocytes, Regulatory/immunology , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Treatment Outcome , Tumor Burden/drug effects
13.
J Clin Apher ; 30(5): 259-64, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25351167

ABSTRACT

Apheresis can remove pathogens and mediators that contribute to pathogenic inflammatory responses in diseases not generally considered to be "Hematologic." Erythrocytapheresis can remove intracellular pathogens such as Babesiosis. Plasmapheresis can remove mediators of the inflammatory response in conditions such as sepsis, chronic autoimmune urticaria and malignant pertussis. Leukapheresis can remove potentially harmful leukocytes in Crohn's Disease and malignant pertussis. While apheresis can remove all of these substances, the clinical efficacy and pathophysiologic changes that occur during apheresis in these conditions are largely unknown. Hence, the clinical utility of apheresis in these conditions is largely unknown and research in these areas has the potential to benefit many patients with a variety of diseases.


Subject(s)
Blood Component Removal , Infections/therapy , Inflammation/therapy , Babesiosis/blood , Babesiosis/parasitology , Babesiosis/therapy , Bacterial Toxins/blood , Chronic Disease , Crohn Disease/blood , Crohn Disease/therapy , Cytokines/blood , Erythrocytes/parasitology , Evidence-Based Medicine , Humans , Infections/blood , Inflammation/blood , Inflammation Mediators/blood , Sepsis/blood , Sepsis/therapy , Urticaria/blood , Urticaria/therapy , Whooping Cough/blood , Whooping Cough/therapy
14.
Clin Cancer Res ; 20(5): 1355-65, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24520093

ABSTRACT

PURPOSE: Myeloma-directed cellular immune responses after autologous stem cell transplantation (ASCT) may reduce relapse rates. We studied whether coinjecting the TLR-3 agonist and vaccine adjuvant Poly-ICLC with a MAGE-A3 peptide vaccine was safe and would elicit a high frequency of vaccine-directed immune responses when combined with vaccine-primed and costimulated autologous T cells. EXPERIMENTAL DESIGN: In a phase II clinical trial (NCT01245673), we evaluated the safety and activity of ex vivo expanded autologous T cells primed in vivo using a MAGE-A3 multipeptide vaccine (compound GL-0817) combined with Poly-ICLC (Hiltonol), granulocyte macrophage colony-stimulating factor (GM-CSF) ± montanide. Twenty-seven patients with active and/or high-risk myeloma received autografts followed by anti-CD3/anti-CD28-costimulated autologous T cells, accompanied by MAGE-A3 peptide immunizations before T-cell collection and five times after ASCT. Immune responses to the vaccine were evaluated by cytokine production (all patients), dextramer binding to CD8(+) T cells, and ELISA performed serially after transplant. RESULTS: T-cell infusions were well tolerated, whereas vaccine injection site reactions occurred in >90% of patients. Two of nine patients who received montanide developed sterile abscesses; however, this did not occur in the 18 patients who did not receive montanide. Dextramer staining demonstrated MAGE-A3-specific CD8 T cells in 7 of 8 evaluable HLA-A2(+) patients (88%), whereas vaccine-specific cytokine-producing T cells were generated in 19 of 25 patients (76%). Antibody responses developed in 7 of 9 patients (78%) who received montanide and only weakly in 2 of 18 patients (11%) who did not. The 2-year overall survival was 74% [95% confidence interval (CI), 54%-100%] and 2-year event-free survival was 56% (95% CI, 37%-85%). CONCLUSIONS: A high frequency of vaccine-specific T-cell responses were generated after transplant by combining costimulated autologous T cells with a Poly-ICLC/GM-CSF-primed MAGE-A3 vaccine.


Subject(s)
Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Immunotherapy, Adoptive , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Neoplasm Proteins/immunology , Poly I-C/immunology , Polylysine/analogs & derivatives , T-Lymphocytes/immunology , Adult , Aged , Cancer Vaccines/immunology , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunization , Immunotherapy, Adoptive/adverse effects , Male , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Polylysine/immunology , Transplantation, Autologous , Treatment Outcome
15.
Cancer ; 120(2): 222-8, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24122387

ABSTRACT

BACKGROUND: Lenalidomide is an immunomodulatory drug with effects on the immune system that may enhance antibody-dependent cell-mediated cytotoxicity and reverse tumor-induced immune suppression. Furthermore, single-agent lenalidomide has therapeutic activity in relapsed/refractory B-cell lymphomas. These immunologic effects potentially may enhance the action of rituximab. METHODS: To test the efficacy of lenalidomide combined with rituximab, the authors conducted a phase 2 trial of lenalidomide, low-dose dexamethasone, and rituximab in patients who had rituximab-resistant, relapsed/refractory, indolent B-cell or mantle cell lymphomas. Patients received two 28-day treatment cycles of lenalidomide 10 mg daily and dexamethasone 8 mg once weekly (part I). During cycle 3, 4 weekly doses of rituximab 375 mg/m2 were administered with lenalidomide-dexamethasone (part II). After the part II response assessment, stable or responding patients continued to receive lenalidomide-dexamethasone. RESULTS: Twenty-seven patients with follicular (n=18), mantle cell (n=5), small lymphocytic (n=3), and marginal zone (n=1) lymphomas started therapy; 3 of 27 patients discontinued therapy because of adverse events and were not evaluable for response. For 24 patients, the overall response rate after part I was 29% (4 patients had a complete response [CR] or CR unconfirmed, and 3 patients had a partial response), and the overall response rate after part II was 58% (8 patients had a CR, and 6 patients had a partial response). For 27 patients, at a median follow-up of 12.2 months, the median progression-free survival was 23.7 months. CONCLUSIONS: The combination of lenalidomide, low-dose dexamethasone, and rituximab achieved high response rates with durable responses in patients with rituximab-resistant, indolent B-cell and mantle cell lymphomas. Overall response rate increased from 29% after two 28-day cycles of lenalidomide and low-dose dexamethasone to 58% after the addition of rituximab, suggesting that lenalidomide can overcome resistance to rituximab.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, B-Cell/drug therapy , Lymphoma, Mantle-Cell/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/administration & dosage , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Lenalidomide , Lymphoma, B-Cell/mortality , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/mortality , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Rituximab , Survival Analysis , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/analogs & derivatives , Treatment Outcome
16.
Hematology Am Soc Hematol Educ Program ; 2014(1): 457-60, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25696894

ABSTRACT

Patients presenting with new or recurrent acute leukemia, particularly of the myeloid lineage, with WBC counts exceeding 100 × 10(9)/L are often considered for leukocytapheresis, especially if they are experiencing symptoms of leukostasis. These symptoms are thought to occur because of blast aggregates and WBC thrombi in the circulation, which reduce blood flow. Leukostasis may cause various complications, including hyperviscosity syndrome, vascular occlusion resulting in intracranial hemorrhages and respiratory failure, and perivascular leukemic infiltrates. Leukostasis occurs more commonly with a high WBC count and with leukemias of monocytoid lineage such as acute myelomonocytic leukemia, which is a reflection of the nature of the leukemic blasts. Leukocytapheresis is used in an effort to quickly decrease a patient's circulating blast count, which can both prevent the development of leukostasis and provide symptomatic relief of leukostasis. However, the impact of leukocytapheresis on early- and long-term mortality is controversial, with several studies producing conflicting results. In this chapter, the pathophysiology of leukostasis, performance of leukocytapheresis, and efficacy of this treatment are reviewed.


Subject(s)
Leukapheresis , Leukemia/complications , Leukocytosis/etiology , Leukocytosis/therapy , Acute Disease , Humans , Plasma Exchange , Treatment Outcome
17.
Mol Cancer Ther ; 11(11): 2321-30, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22964485

ABSTRACT

Despite significant treatment advances over the past decade, multiple myeloma (MM) remains largely incurable. In this study we found that MM cells were remarkably sensitive to the death-inducing effects of a new class of sangivamycin-like molecules (SLM). A panel of structurally related SLMs selectively induced apoptosis in MM cells but not other tumor or nonmalignant cell lines at submicromolar concentrations. SLM6 was the most active compound in vivo, where it was well tolerated and significantly inhibited growth and induced apoptosis of MM tumors. We determined that the anti-MM activity of SLM6 was mediated by direct inhibition of cyclin-dependent kinase 9 (CDK9), which resulted in transcriptional repression of oncogenes that are known to drive MM progression (MAF, CCND1, MYC, and others). Furthermore, SLM6 showed superior in vivo anti-MM activity more than the CDK inhibitor flavopiridol, which is currently in clinical trials for MM. These findings show that SLM6 is a novel CDK9 inhibitor with promising preclinical activity as an anti-MM agent.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Multiple Myeloma/enzymology , Protein Kinase Inhibitors/pharmacology , Pyrimidine Nucleosides/pharmacology , Tubercidin/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/pharmacology , Boronic Acids/therapeutic use , Bortezomib , Cell Death/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , Drug Screening Assays, Antitumor , Flavonoids/pharmacology , Flavonoids/therapeutic use , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Oncogenes , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrazines/pharmacology , Pyrazines/therapeutic use , Pyrimidine Nucleosides/therapeutic use , Translocation, Genetic , Treatment Outcome , Tubercidin/pharmacology , Tubercidin/therapeutic use
18.
Sci Transl Med ; 4(134): 134ra62, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22593175

ABSTRACT

Regulatory T cells (T(regs)) are key mediators of immune tolerance and feature prominently in cancer. Depletion of CD25(+) FoxP3(+) T(regs) in vivo may promote T cell cancer immunosurveillance, but no strategy to do so in humans while preserving immunity and preventing autoimmunity has been validated. We evaluated the Food and Drug Administration-approved CD25-blocking monoclonal antibody daclizumab with regard to human T(reg) survival and function. In vitro, daclizumab did not mediate antibody-dependent or complement-mediated cytotoxicity but rather resulted in the down-regulation of FoxP3 selectively among CD25(high) CD45RA(neg) T(regs). Moreover, daclizumab-treated CD45RA(neg) T(regs) lost suppressive function and regained the ability to produce interferon-γ, consistent with reprogramming. To understand the impact of daclizumab on T(regs) in vivo, we performed a clinical trial of daclizumab in combination with an experimental cancer vaccine in patients with metastatic breast cancer. Daclizumab administration led to a marked and prolonged decrease in T(regs) in patients. Robust CD8 and CD4 T cell priming and boosting to all vaccine antigens were observed in the absence of autoimmunity. We conclude that CD25 blockade depletes and selectively reprograms T(regs) in concert with active immune therapy in cancer patients. These results suggest a mechanism to target cancer-associated T(regs) while avoiding autoimmunity.


Subject(s)
Antibodies, Blocking/therapeutic use , Breast Neoplasms/therapy , Cellular Reprogramming/immunology , Immunotherapy/methods , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Lymphocyte Depletion , T-Lymphocytes, Regulatory/immunology , Antibodies, Blocking/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Combined Modality Therapy , Daclizumab , Female , Forkhead Transcription Factors/metabolism , Humans , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Common Antigens/metabolism , Vaccination
19.
Blood ; 117(1): 63-71, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20864577

ABSTRACT

Severe immune deficiency follows autologous stem cell transplantation for multiple myeloma and is associated with significant infectious morbidity. This study was designed to evaluate the utility of a pretransplantation vaccine and infusion of a primed autologous T-cell product in stimulating specific immunity to influenza. Twenty-one patients with multiple myeloma were enrolled from 2007 to 2009. Patients were randomly assigned to receive an influenza-primed autologous T-cell product or a nonspecifically primed autologous T-cell product. The study endpoint was the development of hemagglutination inhibition titers to the strain-specific serotypes in the influenza vaccine. Enzyme-linked immunospot assays were performed to confirm the development of influenza-specific B-cell and T-cell immunity. Patients who received the influenza-primed autologous T-cell product were significantly more likely to seroconvert in response to the influenza vaccine (P = .001). Seroconversion was accompanied by a significant B-cell response. No differences were observed in the global quantitative recovery of T-cell and B-cell subsets or in global T-cell and B-cell function. The provision of a primed autologous T-cell product significantly improved subsequent influenza vaccine responses. This trial was registered at www.clinicaltrials.gov as #NCT00499577.


Subject(s)
Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Stem Cell Transplantation , T-Lymphocytes/transplantation , Adoptive Transfer , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , Female , Humans , Influenza Vaccines/immunology , Male , Middle Aged , Transplantation, Autologous , Vaccination
20.
Blood ; 117(3): 788-97, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21030558

ABSTRACT

In a phase 1/2 two-arm trial, 54 patients with myeloma received autografts followed by ex vivo anti-CD3/anti-CD28 costimulated autologous T cells at day 2 after transplantation. Study patients positive for human leukocyte antigen A2 (arm A, n = 28) also received pneumococcal conjugate vaccine immunizations before and after transplantation and a multipeptide tumor antigen vaccine derived from the human telomerase reverse transcriptase and the antiapoptotic protein survivin. Patients negative for human leukocyte antigen A2 (arm B, n = 26) received the pneumococcal conjugate vaccine only. Patients exhibited robust T-cell recoveries by day 14 with supraphysiologic T-cell counts accompanied by a sustained reduction in regulatory T cells. The median event-free survival (EFS) for all patients is 20 months (95% confidence interval, 14.6-24.7 months); the projected 3-year overall survival is 83%. A subset of patients in arm A (36%) developed immune responses to the tumor antigen vaccine by tetramer assays, but this cohort did not exhibit better EFS. Higher posttransplantation CD4(+) T-cell counts and a lower percentage of FOXP3(+) T cells were associated with improved EFS. Patients exhibited accelerated polyclonal immunoglobulin recovery compared with patients without T-cell transfers. Adoptive transfer of tumor antigen vaccine-primed and costimulated T cells leads to augmented and accelerated cellular and humoral immune reconstitution, including antitumor immunity, after autologous stem cell transplantation for myeloma. This study was registered at www.clinicaltrials.gov as NCT00499577.


Subject(s)
Immunotherapy/methods , Multiple Myeloma/therapy , Peptide Fragments/immunology , Vaccination/methods , Adult , Aged , Amino Acid Sequence , Antigens, Neoplasm/immunology , Combined Modality Therapy , Exanthema/etiology , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunotherapy/adverse effects , Immunotherapy, Adoptive , Inhibitor of Apoptosis Proteins , Kaplan-Meier Estimate , Male , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/immunology , Middle Aged , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Nausea/etiology , Survivin , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Telomerase/chemistry , Telomerase/immunology , Transplantation, Autologous , Treatment Outcome , Vaccination/adverse effects , Vomiting/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...