1
|
Lavazza C, Budelli S, Montelatici E, Viganò M, Ulbar F, Catani L, Cannone MG, Savelli S, Groppelli E, Lazzari L, Lemoli RM, Cescon M, La Manna G, Giordano R, Montemurro T. Process development and validation of expanded regulatory T cells for prospective applications: an example of manufacturing a personalized advanced therapy medicinal product. J Transl Med 2022; 20:14. [PMID: 34986854 PMCID: PMC8729072 DOI: 10.1186/s12967-021-03200-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A growing number of clinical trials have shown that regulatory T (Treg) cell transfer may have a favorable effect on the maintenance of self-tolerance and immune homeostasis in different conditions such as graft-versus-host disease (GvHD), solid organ transplantation, type 1 diabetes, and others. In this context, the availability of a robust manufacturing protocol that is able to produce a sufficient number of functional Treg cells represents a fundamental prerequisite for the success of a cell therapy clinical protocol. However, extended workflow guidelines for nonprofit manufacturers are currently lacking. Despite the fact that different successful manufacturing procedures and cell products with excellent safety profiles have been reported from early clinical trials, the selection and expansion protocols for Treg cells vary a lot. The objective of this study was to validate a Good Manufacturing Practice (GMP)-compliant protocol for the production of Treg cells that approaches the whole process with a risk-management methodology, from process design to completion of final product development. High emphasis was given to the description of the quality control (QC) methodologies used for the in-process and release tests (sterility, endotoxin test, mycoplasma, and immunophenotype). RESULTS The GMP-compliant protocol defined in this work allows at least 4.11 × 109 Treg cells to be obtained with an average purity of 95.75 ± 4.38% and can be used in different clinical settings to exploit Treg cell immunomodulatory function. CONCLUSIONS These results could be of great use for facilities implementing GMP-compliant cell therapy protocols of these cells for different conditions aimed at restoring the Treg cell number and function, which may slow the progression of certain diseases.
Collapse
Affiliation(s)
- Cristiana Lavazza
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Budelli
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Montelatici
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mariele Viganò
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Ulbar
- Department of Medicine and Aging Sciences, University of Chieti-Pescara, Pescara, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica E Sperimentale, Università di Bologna, Bologna, Italy
| | - Lucia Catani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Dipartimento di Medicina Specialistica, Diagnostica E Sperimentale, Università di Bologna, Bologna, Italy
| | - Marta Giulia Cannone
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sara Savelli
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Groppelli
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenza Lazzari
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberto M Lemoli
- Department of Internal Medicine (DiMI), Clinic of Hematology, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico S. Martino, Genoa, Italy
| | - Matteo Cescon
- Department of General Surgery and Transplantation, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of General Surgery and Transplantation, University of Bologna, Bologna, Italy
| | - Gaetano La Manna
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES)-Nephrology, Dialysis and Renal Transplant Unit, St. Orsola Hospital IRCCS, University of Bologna, Bologna, Italy
| | - Rosaria Giordano
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tiziana Montemurro
- Department of Transfusion Medicine and Hematology, Laboratory of Regenerative Medicine, Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
2
|
Hao Y, Hao S, Andersen-Nissen E, Mauck WM, Zheng S, Butler A, Lee MJ, Wilk AJ, Darby C, Zager M, Hoffman P, Stoeckius M, Papalexi E, Mimitou EP, Jain J, Srivastava A, Stuart T, Fleming LM, Yeung B, Rogers AJ, McElrath JM, Blish CA, Gottardo R, Smibert P, Satija R. Integrated analysis of multimodal single-cell data. Cell 2021; 184:3573-3587.e29. [PMID: 34062119 PMCID: PMC8238499 DOI: 10.1016/j.cell.2021.04.048] [Citation(s) in RCA: 7409] [Impact Index Per Article: 1852.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/03/2021] [Accepted: 04/28/2021] [Indexed: 02/08/2023]
Abstract
The simultaneous measurement of multiple modalities represents an exciting frontier for single-cell genomics and necessitates computational methods that can define cellular states based on multimodal data. Here, we introduce "weighted-nearest neighbor" analysis, an unsupervised framework to learn the relative utility of each data type in each cell, enabling an integrative analysis of multiple modalities. We apply our procedure to a CITE-seq dataset of 211,000 human peripheral blood mononuclear cells (PBMCs) with panels extending to 228 antibodies to construct a multimodal reference atlas of the circulating immune system. Multimodal analysis substantially improves our ability to resolve cell states, allowing us to identify and validate previously unreported lymphoid subpopulations. Moreover, we demonstrate how to leverage this reference to rapidly map new datasets and to interpret immune responses to vaccination and coronavirus disease 2019 (COVID-19). Our approach represents a broadly applicable strategy to analyze single-cell multimodal datasets and to look beyond the transcriptome toward a unified and multimodal definition of cellular identity.
Collapse
Affiliation(s)
- Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Stephanie Hao
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA
| | - Erica Andersen-Nissen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Cape Town HVTN Immunology Lab, Hutchinson Cancer Research Institute of South Africa, Cape Town 8001, South Africa
| | - William M Mauck
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Shiwei Zheng
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Andrew Butler
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Maddie J Lee
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aaron J Wilk
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Charlotte Darby
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Michael Zager
- Center for Data Visualization, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Paul Hoffman
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Marlon Stoeckius
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA
| | - Efthymia Papalexi
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA
| | - Eleni P Mimitou
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA
| | - Jaison Jain
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Avi Srivastava
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Tim Stuart
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA
| | - Lamar M Fleming
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Angela J Rogers
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Juliana M McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94063, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Peter Smibert
- Technology Innovation Lab, New York Genome Center, New York, NY 10013, USA.
| | - Rahul Satija
- Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; New York Genome Center, New York, NY 10013, USA.
| |
Collapse
|
3
|
Elizaga ML, Li SS, Kochar NK, Wilson GJ, Allen MA, Tieu HVN, Frank I, Sobieszczyk ME, Cohen KW, Sanchez B, Latham TE, Clarke DK, Egan MA, Eldridge JH, Hannaman D, Xu R, Ota-Setlik A, McElrath MJ, Hay CM. Safety and tolerability of HIV-1 multiantigen pDNA vaccine given with IL-12 plasmid DNA via electroporation, boosted with a recombinant vesicular stomatitis virus HIV Gag vaccine in healthy volunteers in a randomized, controlled clinical trial. PLoS One 2018; 13:e0202753. [PMID: 30235286 PMCID: PMC6147413 DOI: 10.1371/journal.pone.0202753] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 08/03/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The addition of plasmid cytokine adjuvants, electroporation, and live attenuated viral vectors may further optimize immune responses to DNA vaccines in heterologous prime-boost combinations. The objective of this study was to test the safety and tolerability of a novel prime-boost vaccine regimen incorporating these strategies with different doses of IL-12 plasmid DNA adjuvant. METHODS In a phase 1 study, 88 participants received an HIV-1 multiantigen (gag/pol, env, nef/tat/vif) DNA vaccine (HIV-MAG, 3000 μg) co-administered with IL-12 plasmid DNA adjuvant at 0, 250, 1000, or 1500 μg (N = 22/group) given intramuscularly with electroporation (Ichor TriGrid™ Delivery System device) at 0, 1 and 3 months; followed by attenuated recombinant vesicular stomatitis virus, serotype Indiana, expressing HIV-1 Gag (VSV-Gag), 3.4 ⊆ 107 plaque-forming units (PFU), at 6 months; 12 others received placebo. Injections were in both deltoids at each timepoint. Participants were monitored for safety and tolerability for 15 months. RESULTS The dose of IL-12 pDNA did not increase pain scores, reactogenicity, or adverse events with the co-administered DNA vaccine, or following the VSV-Gag boost. Injection site pain and reactogenicity were common with intramuscular injections with electroporation, but acceptable to most participants. VSV-Gag vaccine often caused systemic reactogenicity symptoms, including a viral syndrome (in 41%) of fever, chills, malaise/fatigue, myalgia, and headache; and decreased lymphocyte counts 1 day after vaccination. CONCLUSIONS HIV-MAG DNA vaccine given by intramuscular injection with electroporation was safe at all doses of IL-12 pDNA. The VSV-Gag vaccine at this dose was associated with fever and viral symptoms in some participants, but the vaccine regimens were safe and generally well-tolerated. TRIAL REGISTRATION Clinical Trials.gov NCT01578889.
Collapse
Affiliation(s)
- Marnie L. Elizaga
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Shuying S. Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Nidhi K. Kochar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Gregory J. Wilson
- Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Mary A. Allen
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hong Van N. Tieu
- Laboratory of Infectious Disease Prevention, New York Blood Center, New York, New York, United States of America
| | - Ian Frank
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Magdalena E. Sobieszczyk
- Division of Infectious Diseases, Columbia University Medical Center, New York, New York, United States of America
| | - Kristen W. Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Brittany Sanchez
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Theresa E. Latham
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - David K. Clarke
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Michael A. Egan
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - John H. Eldridge
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Drew Hannaman
- Ichor Medical Systems, Incorporated, San Diego, California, United States of America
| | - Rong Xu
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - Ayuko Ota-Setlik
- Profectus BioSciences, Incorporated, Pearl River, New York, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Christine Mhorag Hay
- Infectious Diseases Division, University of Rochester Medical Center, Rochester, New York, United States of America
| | | |
Collapse
|
4
|
Margolin K, Morishima C, Velcheti V, Miller JS, Lee SM, Silk AW, Holtan SG, Lacroix AM, Fling SP, Kaiser JC, Egan JO, Jones M, Rhode PR, Rock AD, Cheever MA, Wong HC, Ernstoff MS. Phase I Trial of ALT-803, A Novel Recombinant IL15 Complex, in Patients with Advanced Solid Tumors. Clin Cancer Res 2018; 24:5552-5561. [PMID: 30045932 DOI: 10.1158/1078-0432.ccr-18-0945] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/02/2018] [Accepted: 07/13/2018] [Indexed: 11/16/2022]
Abstract
Purpose: IL15 induces the activation and proliferation of natural killer (NK) and memory CD8+ T cells and has preclinical antitumor activity. Given the superior activity and favorable kinetics of ALT-803 (IL15N72D:IL15RαSu/IgG1 Fc complex) over recombinant human IL15 (rhIL15) in animal models, we performed this first-in-human phase I trial of ALT-803 in patients with advanced solid tumors.Patients and Methods: Patients with incurable advanced melanoma, renal cell, non-small cell lung, and head and neck cancer were treated with ALT-803 0.3 to 6 μg/kg weekly intravenously or 6 to 20 μg/kg weekly subcutaneously for 4 consecutive weeks, every 6 weeks. Immune correlates included pharmacokinetics, immunogenicity, and lymphocyte expansion and function. Clinical endpoints were toxicity and antitumor activity.Results: Twenty-four patients were enrolled; 11 received intravenous and 13 received subcutaneous ALT-803. Of these patients, nine had melanoma, six renal, three head and neck, and six lung cancer. Although total lymphocyte and CD8+ T-cell expansion were modest, NK cell numbers rose significantly. Neither anti-ALT-803 antibodies nor clinical activity were observed. Overall, ALT-803 was well tolerated, with adverse effects including fatigue and nausea most commonly with intravenous administration, whereas painful injection site wheal was reported most commonly with subcutaneous ALT-803.Conclusions: Subcutaneous ALT-803 produced the expected NK cell expansion and was well tolerated with minimal cytokine toxicities and a strong local inflammatory reaction at injection sites in patients with advanced cancer. These data, together with compelling evidence of synergy in preclinical and clinical studies, provide the rationale for combining ALT-803 with other anticancer agents. Clin Cancer Res; 24(22); 5552-61. ©2018 AACR.
Collapse
Affiliation(s)
- Kim Margolin
- City of Hope National Medical Center, Duarte, California.
| | - Chihiro Morishima
- University of Washington, Seattle, Washington.,Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | - Sylvia M Lee
- Seattle Cancer Care Alliance, Seattle, Washington
| | - Ann W Silk
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | | | - Andreanne M Lacroix
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Steven P Fling
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Judith C Kaiser
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jack O Egan
- Altor BioScience, a Nantworks Company, Miramar, Florida
| | - Monica Jones
- Altor BioScience, a Nantworks Company, Miramar, Florida
| | - Peter R Rhode
- Altor BioScience, a Nantworks Company, Miramar, Florida
| | - Amy D Rock
- Altor BioScience, a Nantworks Company, Miramar, Florida
| | - Martin A Cheever
- Cancer Immunotherapy Trials Network, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Hing C Wong
- Altor BioScience, a Nantworks Company, Miramar, Florida
| | | |
Collapse
|
5
|
Schmidt T, Jonat W, Wesch D, Oberg HH, Adam-Klages S, Keller L, Röcken C, Mundhenke C. Influence of physical activity on the immune system in breast cancer patients during chemotherapy. J Cancer Res Clin Oncol 2018; 144:579-586. [PMID: 29305709 DOI: 10.1007/s00432-017-2573-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022]
Abstract
PURPOSE Physical activity can impact the immune system in different ways, e.g. by alteration of the humoral and cellular immune response. Physical activity at medium intensity enhances numbers of cytotoxic T cells, NK cells and macrophages in healthy people. The aim of this study was to compare the effects of endurance and resistance training on the immune system in breast cancer patients during adjuvant chemotherapy. METHODS In a prospective, controlled and randomized intervention exploratory trial, 12-week supervised endurance or resistance training were compared with usual care twice a week. Endpoints were the absolute numbers of the immune cells such as CD3+ T lymphocytes including CD4+- and CD8+, αβ T cells, γδT cells, CD3-/CD16+/56+ NK cells and CD19+ B cells, before and after 12 weeks of treatment. Cell numbers were analyzed using fluorescence-activated cell sorting. RESULTS Despite different physical interventions in all groups immune cell count decreased in CD3 T cells including TCR αβ and CD4 T cells, NK cells and CD19 B cells 12 weeks after initiation of chemotherapy and start of the physical intervention program, while the reduction of γδ T cells and CD8 T cells is less prominent in the RT and UC group. CONCLUSION Chemotherapy led to a decrease in nearly all measured immune cells. In this study, physical intervention with endurance or resistance training did not suppress cellular immunity any further. Larger multicenter trials are needed to evaluate the exact impact of sports intervention on immune cell subpopulations.
Collapse
Affiliation(s)
- Thorsten Schmidt
- Comprehensive Cancer Center North, University of Kiel, Arnold-Heller Straße 3, Haus 14, 24105, Kiel, Germany.
| | - Walter Jonat
- OB/GYN, University of Kiel, Arnold-Heller Straße 3, Haus 24, 24105, Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology, University of Kiel, Arnold-Heller Straße 3, 24105, Kiel, Germany
| | - Hans-Heinrich Oberg
- Institute of Immunology, University of Kiel, Arnold-Heller Straße 3, 24105, Kiel, Germany
| | - Sabine Adam-Klages
- Institute of Immunology, University of Kiel, Arnold-Heller Straße 3, 24105, Kiel, Germany
| | - Lisa Keller
- OB/GYN, University of Kiel, Arnold-Heller Straße 3, Haus 24, 24105, Kiel, Germany
| | - Christoph Röcken
- Comprehensive Cancer Center North, University of Kiel, Arnold-Heller Straße 3, Haus 14, 24105, Kiel, Germany
| | - Christoph Mundhenke
- OB/GYN, University of Kiel, Arnold-Heller Straße 3, Haus 24, 24105, Kiel, Germany
| |
Collapse
|
6
|
Miller JS, Morishima C, McNeel DG, Patel MR, Kohrt HEK, Thompson JA, Sondel PM, Wakelee HA, Disis ML, Kaiser JC, Cheever MA, Streicher H, Creekmore SP, Waldmann TA, Conlon KC. A First-in-Human Phase I Study of Subcutaneous Outpatient Recombinant Human IL15 (rhIL15) in Adults with Advanced Solid Tumors. Clin Cancer Res 2017; 24:1525-1535. [PMID: 29203590 DOI: 10.1158/1078-0432.ccr-17-2451] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/22/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022]
Abstract
Purpose: Preclinical data established IL15 as a homeostatic factor and powerful stimulator of NK and CD8+ T-cell function, the basis for clinical testing.Experimental Design: A first-in-human outpatient phase I dose escalation trial of subcutaneous (SC) rhIL15 was conducted in refractory solid tumor cancer patients. Therapy consisted of daily (Monday-Friday) subcutaneous injections of rhIL15 for two consecutive weeks (10 total doses/cycle). Clinical response was assessed by RECIST. Pharmacokinetics of rhIL15 and immune biomarkers were evaluated.Results: Nineteen patients were treated with rhIL15 at dose levels of 0.25, 0.5, 1, 2, and 3 mcg/kg/day. Fourteen patients completed ≥ 2 cycles of therapy that was well tolerated. One serious adverse event (SAE), grade 2 pancreatitis, required overnight hospitalization. Enrollment was halted after a patient receiving 3 mcg/kg/day developed a dose-limiting SAE of grade 3 cardiac chest pain associated with hypotension and increased troponin. No objective responses were observed; however, several patients had disease stabilization including a renal cell carcinoma patient who continued protocol treatment for 2 years. The treatment induced profound expansion of circulating NK cells, especially among the CD56bright subset. A proportional but less dramatic increase was found among circulating CD8+ T cells with maximal 3-fold expansion for the 2 and 3 mcg/kg patients.Conclusions: SC rhIL15 treatment was well tolerated, producing substantial increases in circulating NK and CD8+ T cells. This protocol establishes a safe outpatient SC rhIL15 regimen of 2 mcg/kg/day dosing amenable to self-injection and with potential as a combination immunotherapeutic agent. Clin Cancer Res; 24(7); 1525-35. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | - John A Thompson
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,National Cancer Institute/NIH, Bethesda, Maryland
| | | | | | | | | | | | - Howard Streicher
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,National Cancer Institute/NIH, Bethesda, Maryland
| | - Steven P Creekmore
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,National Cancer Institute/NIH, Bethesda, Maryland
| | - Thomas A Waldmann
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,National Cancer Institute/NIH, Bethesda, Maryland
| | - Kevin C Conlon
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,National Cancer Institute/NIH, Bethesda, Maryland
| |
Collapse
|
7
|
Arber DA, Borowitz MJ, Cessna M, Etzell J, Foucar K, Hasserjian RP, Rizzo JD, Theil K, Wang SA, Smith AT, Rumble RB, Thomas NE, Vardiman JW. Initial Diagnostic Workup of Acute Leukemia: Guideline From the College of American Pathologists and the American Society of Hematology. Arch Pathol Lab Med 2017; 141:1342-1393. [PMID: 28225303 DOI: 10.5858/arpa.2016-0504-cp] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - A complete diagnosis of acute leukemia requires knowledge of clinical information combined with morphologic evaluation, immunophenotyping and karyotype analysis, and often, molecular genetic testing. Although many aspects of the workup for acute leukemia are well accepted, few guidelines have addressed the different aspects of the diagnostic evaluation of samples from patients suspected to have acute leukemia. OBJECTIVE - To develop a guideline for treating physicians and pathologists involved in the diagnostic and prognostic evaluation of new acute leukemia samples, including acute lymphoblastic leukemia, acute myeloid leukemia, and acute leukemias of ambiguous lineage. DESIGN - The College of American Pathologists and the American Society of Hematology convened a panel of experts in hematology and hematopathology to develop recommendations. A systematic evidence review was conducted to address 6 key questions. Recommendations were derived from strength of evidence, feedback received during the public comment period, and expert panel consensus. RESULTS - Twenty-seven guideline statements were established, which ranged from recommendations on what clinical and laboratory information should be available as part of the diagnostic and prognostic evaluation of acute leukemia samples to what types of testing should be performed routinely, with recommendations on where such testing should be performed and how the results should be reported. CONCLUSIONS - The guideline provides a framework for the multiple steps, including laboratory testing, in the evaluation of acute leukemia samples. Some aspects of the guideline, especially molecular genetic testing in acute leukemia, are rapidly changing with new supportive literature, which will require on-going updates for the guideline to remain relevant.
Collapse
|
8
|
Koguchi Y, Gonzalez IL, Meeuwsen TL, Miller WL, Haley DP, Tanibata-Branham AN, Bahjat KS. A Semi-automated Approach to Preparing Antibody Cocktails for Immunophenotypic Analysis of Human Peripheral Blood. J Vis Exp 2016:e53485. [PMID: 26890325 PMCID: PMC4781742 DOI: 10.3791/53485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Immunophenotyping of peripheral blood by flow cytometry determines changes in the frequency and activation status of peripheral leukocytes during disease and treatment. It has the potential to predict therapeutic efficacy and identify novel therapeutic targets. Whole blood staining utilizes unmanipulated blood, which minimizes artifacts that can occur during sample preparation. However, whole blood staining must also be done on freshly collected blood to ensure the integrity of the sample. Additionally, it is best to prepare antibody cocktails on the same day to avoid potential instability of tandem-dyes and prevent reagent interaction between brilliant violet dyes. Therefore, whole blood staining requires careful standardization to control for intra and inter-experimental variability. Here, we report deployment of an automated liquid handler equipped with a two-dimensional (2D) barcode reader into a standard process of making antibody cocktails for flow cytometry. Antibodies were transferred into 2D barcoded tubes arranged in a 96 well format and their contents compiled in a database. The liquid handler could then locate the source antibody vials by referencing antibody names within the database. Our method eliminated tedious coordination for positioning of source antibody tubes. It provided versatility allowing the user to easily change any number of details in the antibody dispensing process such as specific antibody to use, volume, and destination by modifying the database without rewriting the scripting in the software method for each assay. A proof of concept experiment achieved outstanding inter and intra- assay precision, demonstrated by replicate preparation of an 11-color, 17-antibody flow cytometry assay. These methodologies increased overall throughput for flow cytometry assays and facilitated daily preparation of the complex antibody cocktails required for the detailed phenotypic characterization of freshly collected anticoagulated peripheral blood.
Collapse
Affiliation(s)
- Yoshinobu Koguchi
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center;
| | - Iliana L Gonzalez
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center
| | - Tanisha L Meeuwsen
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center
| | - William L Miller
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center
| | - Daniel P Haley
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center; Sony Biotechnology
| | | | - Keith S Bahjat
- Human Immune Monitoring Laboratory, Earle A. Chiles Research Institute, Providence Cancer Center, Providence Portland Medical Center; Bristol-Myers Squibb;
| |
Collapse
|
9
|
Oberg HH, Kellner C, Peipp M, Sebens S, Adam-Klages S, Gramatzki M, Kabelitz D, Wesch D. Monitoring Circulating γδ T Cells in Cancer Patients to Optimize γδ T Cell-Based Immunotherapy. Front Immunol 2014; 5:643. [PMID: 25566256 PMCID: PMC4269191 DOI: 10.3389/fimmu.2014.00643] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/03/2014] [Indexed: 12/25/2022] Open
Abstract
The success of γδ T cell-based immunotherapy, where the cytotoxic activity of circulating γδ T lymphocytes is activated by nitrogen-containing bisphosphonates (n-BP), or possibly by bispecific antibodies or the combination of both, requires a profound knowledge of patients' γδ T cells. A possible influence of radio- or chemotherapy on γδ T cells as well as their reported exhaustion after repetitive treatment with n-BP or their lack of response to various cancers can be easily determined by the monitoring assays described in this perspective article. Monitoring the absolute cell numbers of circulating γδ T cell subpopulations in small volumes of whole blood from cancer patients and determining γδ T cell cytotoxicity using the Real-Time Cell Analyzer can give a more comprehensive assessment of a personalized tumor treatment. Possible future directions such as the combined usage of n-BP or phosphorylated antigens together with bispecific antibodies that selectively target γδ T cells to tumor-associated antigens, will be discussed. Such strategies induce expansion and enhance γδ T cell cytotoxicity and might possibly avoid their exhaustion and overcome the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Hans-Heinrich Oberg
- Institute of Immunology, Christian-Albrechts-University of Kiel , Kiel , Germany
| | - Christian Kellner
- 2nd Medical Department, Division of Stem Cell Transplantation and Immunotherapy, Christian-Albrechts-University of Kiel , Kiel , Germany
| | - Matthias Peipp
- 2nd Medical Department, Division of Stem Cell Transplantation and Immunotherapy, Christian-Albrechts-University of Kiel , Kiel , Germany
| | - Susanne Sebens
- Institute for Experimental Medicine, Christian-Albrechts-University of Kiel , Kiel , Germany
| | - Sabine Adam-Klages
- Institute of Immunology, Christian-Albrechts-University of Kiel , Kiel , Germany
| | - Martin Gramatzki
- 2nd Medical Department, Division of Stem Cell Transplantation and Immunotherapy, Christian-Albrechts-University of Kiel , Kiel , Germany
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University of Kiel , Kiel , Germany
| | - Daniela Wesch
- Institute of Immunology, Christian-Albrechts-University of Kiel , Kiel , Germany
| |
Collapse
|
10
|
Hensley-McBain T, Heit A, De Rosa SC, McElrath MJ, Andersen-Nissen E. Optimization of a whole blood phenotyping assay for enumeration of peripheral blood leukocyte populations in multicenter clinical trials. J Immunol Methods 2014; 411:23-36. [PMID: 24925805 DOI: 10.1016/j.jim.2014.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 05/28/2014] [Accepted: 06/02/2014] [Indexed: 10/25/2022]
Abstract
Vaccination with viral vectors or adjuvants can induce early changes in circulating peripheral blood leukocytes that are predictive of a protective immune response. In this study, we define an 11-color whole blood antibody staining Trucount Panel (TP1) to enumerate and phenotype the major leukocyte populations in a human vaccine experimental medicine trial setting. TP1 can be prepared up to 8weeks prior to use, enabling bulk preparation at a central laboratory and distribution to clinical sites. Cells in whole blood must be stained within 4h of draw to accurately detect the major cell populations. Staining of cells with TP1 followed by storage and shipping at -80°C to a central laboratory has little to no effect on the cell concentrations observed. We also present data from an HIV vaccine multicenter clinical trial obtained using the optimized TP1 assay protocol and show that the data produced accurately correlates with complete blood count (CBC) data. Taken together, these data indicate the optimized TP1 panel assay can be used in a multicenter clinical trial setting to increase our understanding of systemic responses to vaccination or disease.
Collapse
Affiliation(s)
- Tiffany Hensley-McBain
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Antje Heit
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Stephen C De Rosa
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | - M Juliana McElrath
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Erica Andersen-Nissen
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Cape Town HVTN Immunology Laboratory, Hutchinson Center Research Institute of South Africa, Cape Town, South Africa.
| |
Collapse
|
11
|
Seshadri C, Shenoy M, Wells RD, Hensley-McBain T, Andersen-Nissen E, McElrath MJ, Cheng TY, Moody DB, Hawn TR. Human CD1a deficiency is common and genetically regulated. THE JOURNAL OF IMMUNOLOGY 2013; 191:1586-93. [PMID: 23858036 DOI: 10.4049/jimmunol.1300575] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
CD1 proteins evolved to present diverse lipid Ags to T cells. In comparison with MHC proteins, CD1 proteins exhibit minimal allelic diversity as a result of nonsynonymous single nucleotide polymorphisms (SNPs). However, it is unknown if common SNPs in gene regulatory regions affect CD1 expression and function. We report surprising diversity in patterns of inducible CD1a expression on human dendritic cells (DCs), spanning the full range from undetectable to high density, a finding not seen with other CD1 isoforms. CD1a-deficient DCs failed to present mycobacterial lipopeptide to T cells but had no defects in endocytosis, cytokine secretion, or expression of costimulatory molecules after LPS treatment. We identified an SNP in the 5' untranslated region (rs366316) that was common and strongly associated with low CD1a surface expression and mRNA levels (p = 0.03 and p = 0.001, respectively). Using a CD1a promoter-luciferase system in combination with mutagenesis studies, we found that the polymorphic allele reduced luciferase expression by 44% compared with the wild-type variant (p < 0.001). Genetic regulation of lipid Ag presentation by varying expression on human DCs provides a mechanism for achieving population level differences in immune responses despite limited structural variation in CD1a proteins.
Collapse
Affiliation(s)
- Chetan Seshadri
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|