1
|
Keruakous AR, Asch A, Aljumaily R, Zhao D, Yuen C. Prognostic impact of natural killer cell recovery on minimal residual disease after autologous stem cell transplantation in multiple myeloma. Transpl Immunol 2022; 71:101544. [PMID: 35093506 PMCID: PMC10434759 DOI: 10.1016/j.trim.2022.101544] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/20/2022] [Accepted: 01/23/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Natural killer cells are a potent effector lymphocyte subset that can induce cytotoxicity without the need for antigen sensitization or presentation. NK cells are a tempting target -for immune therapy, monoclonal antibody, or genetic engineering-to enhance immune surveillance mechanisms against myeloma cells. MATERIALS AND METHODS We hypothesized an association between natural killer cell recovery after autologous stem cell transplantation (ASCT) and disease outcomes in multiple myeloma patients. We concluded a prospective study that started enrolling patients in January 2020 to identify the association between absolute NK cell count two to three after ASCT and disease outcomes after autologous stem cell transplantation in multiple myeloma using univariate and multivariate analysis. RESULTS Natural killer cell recovery was evaluated during the third month after ASCT, day +60 to +90 post-ASCT. Our patients had a mean NK cell count of 90.53, ranging from 14 to 282 Cell/μL (Std Dev 84.64 Cell/μL). The odds of having a minimal residual disease (MRD-positivity) among patients with partial remission before transplantation is four times higher than patients with very good partial response or better (95% confidence interval 0.45-35.79). Our patients were classified into two groups based on MRD status after ASCT, an MRD-negative group of eight participants and an MRD-positive group of seven participants. The mean absolute NK cell count was significantly higher in the MRD-negative cohort, 131.38 Cell/μL, versus 43.86 Cell/μL in the MRD-positive group (p = 0.049). CONCLUSION We conclude that for multiple myeloma patients treated with ASCT, high absolute NK cell counts two to three months after ASCT is an independent predictor for MRD negativity.
Collapse
Affiliation(s)
- Amany R Keruakous
- Georgia Cancer Center at Augusta University, Augusta, GA, United States of America.
| | - Adam Asch
- Department of Hematology/Oncology, University of Oklahoma, Oklahoma City, OK, United States of America
| | - Raid Aljumaily
- Department of Hematology/Oncology, University of Oklahoma, Oklahoma City, OK, United States of America
| | - Daniel Zhao
- Department of Biostatistics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Carrie Yuen
- Department of Hematology/Oncology, University of Oklahoma, Oklahoma City, OK, United States of America
| |
Collapse
|
2
|
Mohanlal R, Qiu Y, Zheng M, Mirkou A, Sridharan K, Keir C. Long-Term Safety Follow-Up of Subjects Previously Treated with Non-Replicating Retroviral Vector-Based Gene Therapies. Mol Diagn Ther 2017; 20:591-602. [PMID: 27435702 PMCID: PMC5107205 DOI: 10.1007/s40291-016-0229-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective Our objective was to evaluate the life-long safety profile of gene therapy using retroviral (non-replicating) vectors (nRCR), or cell products in 127 subjects with hemophilia, human immunodeficiency virus (HIV), or cancer, previously treated with such gene therapy. Methods We assessed the occurrence of serious adverse events (SAEs), deaths and presence of replication competent retrovirus (RCR). Results A total of 23 subjects remained until the data cut-off date of 31 July 2013 and provided safety information of up to 18 years. Of the 104 subjects who discontinued, the primary reason was loss to follow-up (47.2 %; n = 60). The follow-up period for the 60 subjects lost to follow-up was 7–10 years. A total of 41 subjects experienced at least one SAE, and 15 subjects died. We reviewed SAEs and cause of death (none related to the active therapy), but no evidence was found for safety signals related to new malignancy or neurologic, rheumatological, autoimmune, or hematologic disorder. RCR results were negative, indicating no evidence for in vivo vector persistence. Conclusion Despite the loss of follow-up, which is the limiting factor in this long-term safety trial, the findings from this long-term follow-up study are encouraging.
Collapse
Affiliation(s)
- Ramon Mohanlal
- Novartis Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Yuhong Qiu
- Novartis Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Ming Zheng
- Novartis Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Asmae Mirkou
- Novartis Oncology, Novartis Pharma AG, Basel, Switzerland
| | - Kanaka Sridharan
- Cell and Gene Therapies Unit, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Christopher Keir
- Cell and Gene Therapies Unit, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| |
Collapse
|
3
|
Hardy CL. Specificity of Hematopoietic Stem and Progenitor Cell Homing to Bone Marrow: A Perspective. Hematology 2016; 5:391-401. [DOI: 10.1080/10245332.2000.11746535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Cheryl L. Hardy
- G.V. (Sonny) Montgomery Department of Veterans Affairs Medical Center, Department of Medicine, University of Mississippi School of Medicine, Jackson, MS
| |
Collapse
|
4
|
Smith JA, Goldspiel BR. Cancer gene therapy update. J Oncol Pharm Pract 2016. [DOI: 10.1177/107815529900500101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective. To provide an update about gene marking and gene therapy trials in cancer patients. Data Sources. A MEDLINE search using the term “gene therapy” was conducted for the period 1985 to 1998. The reference lists from retrieved articles were reviewed. Meeting abstracts from the American Society of Clinical Oncology annual meeting (published in their proceedings) and the Annual Cancer Gene Therapy Symposium (published in Cancer Gene Therapy) that concerned gene therapy in cancer patients were also included. Data Extraction. Both authors reviewed the retrieved material and included preclinical data, case reports, and clinical trials related to gene transfer or gene therapy in cancer patients. Data Synthesis. There are several possible approaches to using gene therapy for the diagnosis and treatment of cancer and for the monitoring of cancer therapy. Exogenous genes may be used to mark cells to help better understand cancer biology or may be used directly for cancer treatment. Gene-marking trials have already provided new information about cancer biology and have demonstrated that reinfused progenitor cells may be a source of relapse in patients with acute or chronic myelogenous leukemia and neuroblastoma. Approaches using gene therapy for cancer treatment include: using lymphocytes as gene carriers, using foreign genes to increase tumor immunogenicity, introducing tumor regression antigen genes into viruses, introducing “sensitivity” genes to produce new cytotoxic agent(s) within tumors, producing new protein product(s) to protect normal cells, replacing missing or mutant tumor suppressor genes, and inactivating oncogenes. Clinical trials using these strategies have demonstrated that gene transfer is feasible (albeit with low transduction efficiency) and that gene expression occurs; in addition, clinical responses have been noted.
Collapse
Affiliation(s)
- Judith A Smith
- National Institutes of Health Clinical Center Pharmacy Department, Bethesda, Maryland
| | - Barry R Goldspiel
- National Institutes of Health Clinical Center Pharmacy Department, Bethesda, Maryland
| |
Collapse
|
5
|
Taverna JA, Yun S, Jonnadula J, Saleh A, Riaz IB, Abraham I, Yeager AM, Persky DO, McBride A, Haldar S, Anwer F. Role of Maintenance Therapy after High-Dose Chemotherapy and Autologous Hematopoietic Cell Transplantation in Aggressive Lymphomas: A Systematic Review. Biol Blood Marrow Transplant 2016; 22:1182-1196. [PMID: 26899562 DOI: 10.1016/j.bbmt.2016.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/08/2016] [Indexed: 11/29/2022]
Abstract
Significant uncertainty exists in regard to the efficacy of maintenance therapy after high-dose chemotherapy (HDC) as well as autologous stem cell transplantation (ASCT) for the treatment of patients with aggressive lymphoma. A systematic review was performed to evaluate the effectiveness of post-ASCT maintenance therapy in patients with relapsed/refractory lymphoma. A comprehensive literature search yielded 4476 studies and a total of 42 studies (11 randomized controlled trials [RCT], 9 retrospective comparative studies, and 22 single-arm studies) were included in the systematic review. There was significant heterogeneity in study design, chemotherapeutic regimens, post-ASCT maintenance strategies, patient enrollment criteria, and study endpoints. Our findings suggest that post-ASCT maintenance immune-targeting strategies, including PD-1/PD-L1 blocking antibodies, rituximab, and brentuximab, may improve progression-free survival but not overall survival. Collectively, the results indicate a need for testing new strategies with well-designed and adequately powered RCTs to better address the role of post-ASCT maintenance in relapsed/refractory lymphomas.
Collapse
Affiliation(s)
- Josephine A Taverna
- Department of Medicine, University of Arizona, Tucson, Arizona; Department of Hematology and Oncology, Blood and Marrow Transplantation Program, University of Arizona, Tucson, Arizona; Department of Hematology and Oncology, Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Seongseok Yun
- Department of Medicine, University of Arizona, Tucson, Arizona
| | | | - Ahlam Saleh
- Arizona Health Sciences Library, University of Arizona, Tucson, Arizona
| | - Irbaz Bin Riaz
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Ivo Abraham
- Center for Health Outcomes and PharmacoEconomic Research, University of Arizona, Tucson, Arizona; Arizona Cancer Center, Tucson, Arizona
| | - Andrew M Yeager
- Department of Medicine, University of Arizona, Tucson, Arizona; Department of Hematology and Oncology, Blood and Marrow Transplantation Program, University of Arizona, Tucson, Arizona; Arizona Cancer Center, Tucson, Arizona
| | - Daniel O Persky
- Department of Medicine, University of Arizona, Tucson, Arizona; Department of Hematology and Oncology, Blood and Marrow Transplantation Program, University of Arizona, Tucson, Arizona; Arizona Cancer Center, Tucson, Arizona
| | - Ali McBride
- Department of Hematology and Oncology, Blood and Marrow Transplantation Program, University of Arizona, Tucson, Arizona; Arizona Cancer Center, Tucson, Arizona
| | - Subrata Haldar
- Department of Hematology and Oncology, Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Faiz Anwer
- Department of Medicine, University of Arizona, Tucson, Arizona; Department of Hematology and Oncology, Blood and Marrow Transplantation Program, University of Arizona, Tucson, Arizona; Arizona Cancer Center, Tucson, Arizona.
| |
Collapse
|
6
|
Larochelle A, Dunbar CE. Hematopoietic stem cell gene therapy:assessing the relevance of preclinical models. Semin Hematol 2014; 50:101-30. [PMID: 24014892 DOI: 10.1053/j.seminhematol.2013.03.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
7
|
Comparison of Autologous Hematopoietic Cell Transplantation and Chemotherapy as Postremission Treatment in Non-M3 Acute Myeloid Leukemia in First Complete Remission. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:444-51. [DOI: 10.1016/j.clml.2012.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 06/19/2012] [Accepted: 07/26/2012] [Indexed: 11/19/2022]
|
8
|
Schuurhuis GJ, Zweegman S, Ossenkoppele GJ. The adverse prognosis for acute myeloid leukemia patients with superior autologous stem cell mobilization. Leuk Res 2012; 36:1323-4. [DOI: 10.1016/j.leukres.2012.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 07/02/2012] [Accepted: 07/07/2012] [Indexed: 10/27/2022]
|
9
|
Abstract
The genetic engineering of hematopoietic stem cells is the basis for potentially treating a large array of hereditary and acquired diseases, and stands as the paradigm for stem cell engineering in general. Recent clinical reports support the formidable promise of this approach but also highlight the limitations of the technologies used to date, which have on occasion resulted in clonal expansion, myelodysplasia, or leukemogenesis. New research directions, predicated on improved vector designs, targeted gene delivery or the therapeutic use of pluripotent stem cells, herald the advent of safer and more effective hematopoietic stem cell therapies that may transform medical practice. In this review, we place these recent advances in perspective, emphasizing the solutions emerging from a wave of new technologies and highlighting the challenges that lie ahead.
Collapse
|
10
|
DiPersio JF, Ho AD, Hanrahan J, Hsu FJ, Fruehauf S. Relevance and Clinical Implications of Tumor Cell Mobilization in the Autologous Transplant Setting. Biol Blood Marrow Transplant 2011; 17:943-55. [DOI: 10.1016/j.bbmt.2010.10.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 10/15/2010] [Indexed: 01/04/2023]
|
11
|
Barese CN, Dunbar CE. Contributions of gene marking to cell and gene therapies. Hum Gene Ther 2011; 22:659-68. [PMID: 21261461 DOI: 10.1089/hum.2010.237] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The first human genetic modification studies used replication-incompetent integrating vector vectors to introduce marker genes into T lymphocytes and subsequently into hematopoietic stem cells. Such studies have provided numerous insights into the biology of hematopoiesis and immune reconstitution and contributed to clinical development of gene and cell therapies. Tracking of hematopoietic reconstitution and analysis of the origin of residual malignant disease after hematopoietic transplantation has been possible via gene marking. Introduction of selectable marker genes has enabled preselection of specific T-cell populations for tumor and viral immunotherapy and reduced the threat of graft-versus-host disease, improving the survival of patients after allogeneic marrow transplantation. Marking studies in humans, murine xenografts, and large animals have helped optimize conditions for gene transfer into CD34(+) hematopoietic progenitors, contributing to the achievement of gene transfer efficiencies sufficient for clinical benefit in several serious genetic diseases such as X-linked severe combined immunodeficiency and adrenoleukodystrophy. When adverse events linked to insertional mutagenesis arose in clinical gene therapy trials for inherited immunodeficiencies, additional animal studies using gene-marking vectors have greatly increased our understanding of genotoxicity. The knowledge gained from these studies is being translated into new vector designs and clinical protocols, which we hope will continue to improve the efficiency, effectiveness and safety of these promising therapeutic approaches.
Collapse
Affiliation(s)
- Cecilia N Barese
- Hematology Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20852, USA
| | | |
Collapse
|
12
|
Carrasco JL. A generalized concordance correlation coefficient based on the variance components generalized linear mixed models for overdispersed count data. Biometrics 2011; 66:897-904. [PMID: 19817740 DOI: 10.1111/j.1541-0420.2009.01335.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The classical concordance correlation coefficient (CCC) to measure agreement among a set of observers assumes data to be distributed as normal and a linear relationship between the mean and the subject and observer effects. Here, the CCC is generalized to afford any distribution from the exponential family by means of the generalized linear mixed models (GLMMs) theory and applied to the case of overdispersed count data. An example of CD34+ cell count data is provided to show the applicability of the procedure. In the latter case, different CCCs are defined and applied to the data by changing the GLMM that fits the data. A simulation study is carried out to explore the behavior of the procedure with a small and moderate sample size.
Collapse
Affiliation(s)
- Josep L Carrasco
- Bioestadistica, Departament de Salut Publica, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
13
|
Abstract
Large animal models have been instrumental in advancing hematopoietic stem cell (HSC) gene therapy. Here we review the advantages of large animal models, their contributions to the field of HSC gene therapy and recent progress in this field. Several properties of human HSCs including their purification, their cell-cycle characteristics, their response to cytokines and the proliferative demands placed on them after transplantation are more similar in large animal models than in mice. Progress in the development and use of retroviral vectors and ex vivo transduction protocols over the last decade has led to efficient gene transfer in both dogs and nonhuman primates. Importantly, the approaches developed in these models have translated well to the clinic. Large animals continue to be useful to evaluate the efficacy and safety of gene therapy, and dogs with hematopoietic diseases have now been cured by HSC gene therapy. Nonhuman primates allow evaluation of aspects of transplantation as well as disease-specific approaches such as AIDS (acquired immunodeficiency syndrome) gene therapy that can not be modeled well in the dog. Finally, large animal models have been used to evaluate the genotoxicity of viral vectors by comparing integration sites in hematopoietic repopulating cells and monitoring clonality after transplantation.
Collapse
Affiliation(s)
- G D Trobridge
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | | |
Collapse
|
14
|
Escors D, Breckpot K. Lentiviral vectors in gene therapy: their current status and future potential. Arch Immunol Ther Exp (Warsz) 2010; 58:107-19. [PMID: 20143172 DOI: 10.1007/s00005-010-0063-4] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 10/06/2009] [Indexed: 12/28/2022]
Abstract
The concept of gene therapy originated in the mid twentieth century and was perceived as a revolutionary technology with the promise to cure almost any disease of which the molecular basis was understood. Since then, several gene vectors have been developed and the feasibility of gene therapy has been shown in many animal models of human disease. However, clinical efficacy could not be demonstrated until the beginning of the new century in a small-scale clinical trial curing an otherwise fatal immunodeficiency disorder in children. This first success, achieved after retroviral therapy, was later overshadowed by the occurrence of vector-related leukemia in a significant number of the treated children, demonstrating that the future success of gene therapy depends on our understanding of vector biology. This has led to the development of later-generation vectors with improved efficiency, specificity, and safety. Amongst these are HIV-1 lentivirus-based vectors (lentivectors), which are being increasingly used in basic and applied research. Human gene therapy clinical trials are currently underway using lentivectors in a wide range of human diseases. The intention of this review is to describe the main scientific steps leading to the engineering of HIV-1 lentiviral vectors and place them in the context of current human gene therapy.
Collapse
Affiliation(s)
- David Escors
- Division of Infection and Immunity, Medical School of the Royal Free and University College London, London W1T 4JF, UK.
| | | |
Collapse
|
15
|
Persons DA. Hematopoietic stem cell gene transfer for the treatment of hemoglobin disorders. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2009; 2009:690-697. [PMID: 20008255 DOI: 10.1182/asheducation-2009.1.690] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Hematopoietic stem cell (HSC)-targeted gene transfer is an attractive approach for the treatment of a number of hematopoietic disorders caused by single gene defects. Indeed, in a series of gene transfer trials for two different primary immunodeficiencies beginning early in this decade, outstanding success has been achieved. Despite generally low levels of engrafted, genetically modified HSCs, these trials were successful because of the marked selective advantage of gene-corrected lymphoid precursors that allowed reconstitution of the immune system. Unlike the immunodeficiencies, this robust level of in vivo selection is not available to hematopoietic repopulating cells or early progenitor cells following gene transfer of a therapeutic globin gene in the setting of beta-thalassemia and sickle cell disease. Both preclinical and clinical transplant studies involving bone marrow chimeras suggest that 20% or higher levels of engraftment of genetically modified HSCs will be needed for clinical success in the most severe of these disorders. Encouragingly, gene transfer levels in this range have recently been reported in a lentiviral vector gene transfer clinical trial for children with adrenoleukodystrophy. A clinical gene transfer trial for beta-thalassemia has begun in France, and one patient with transfusion-dependent HbE/beta-thalassemia has demonstrated a therapeutic effect after transplantation with autologous CD34(+) cells genetically modified with a beta-globin lentiviral vector. Here, the development and recent progress of gene therapy for the hemoglobin disorders is reviewed.
Collapse
Affiliation(s)
- Derek A Persons
- Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
16
|
Abstract
The concept of introducing genes into human cells for therapeutic purposes developed nearly 50 years ago as diseases due to defects in specific genes were recognized. Development of recombinant DNA techniques in the 1970s and their application to the study of mouse tumor viruses facilitated the assembly of the first gene transfer vectors. Vectors of several different types have now been developed for specific applications and over the past decade, efficacy has been demonstrated in many animal models. Clinical trials began in 1989 and by 2002 there was unequivocal evidence that children with severe combined immunodeficiency could be cured by gene transfer into primitive hematopoietic cells. Emerging from these successful trials was the realization that proto-oncogene activation by retroviral integration could contribute to leukemia. Much current effort is focused on development of safer vectors. Successful gene therapy applications have also been developed for control of graft-versus-host disease and treatment of various viral infections, leukemias, and lymphomas. The hemophilias seem amenable to gene therapy intervention and informative clinical trials have been conducted. The hemoglobin disorders, an early target for gene therapy, have proved particularly challenging although ongoing research is yielding new information that may ultimately lead to successful clinical trials.
Collapse
|
17
|
Abstract
There is a critical need to develop new and effective cancer therapies that target bone, the primary metastatic site for prostate cancer and other malignancies. Among the various therapeutic approaches being considered for this application, gene-modified cell-based therapies may have specific advantages. Gene-modified cell therapy uses gene transfer and cell-based technologies in a complementary fashion to chaperone appropriate gene expression cassettes to active sites of tumor growth. In this paper, we briefly review potential cell vehicles for this approach and discuss relevant gene therapy strategies for prostate cancer. We further discuss selected studies that led to the conceptual development and preclinical testing of IL-12 gene-modified bone marrow cell therapy for prostate cancer. Finally, we discuss future directions in the development of gene-modified cell therapy for metastatic prostate cancer, including the need to identify and test novel therapeutic genes such as GLIPR1.
Collapse
Affiliation(s)
- H Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
18
|
Deola S, Scaramuzza S, Birolo RS, Cergnul M, Ficara F, Dando J, Voena C, Vai S, Monari M, Pogliani E, Corneo G, Peccatori J, Selleri S, Bordignon C, Roncarolo MG, Aiuti A, Bregni M. Molecular purging of multiple myeloma cells by ex-vivo culture and retroviral transduction of mobilized-blood CD34+ cells. J Transl Med 2007; 5:35. [PMID: 17626627 PMCID: PMC1948885 DOI: 10.1186/1479-5876-5-35] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 07/12/2007] [Indexed: 11/21/2022] Open
Abstract
Background Tumor cell contamination of the apheresis in multiple myeloma is likely to affect disease-free and overall survival after autografting. Objective To purge myeloma aphereses from tumor contaminants with a novel culture-based purging method. Methods We cultured myeloma-positive CD34+ PB samples in conditions that retained multipotency of hematopoietic stem cells, but were unfavourable to survival of plasma cells. Moreover, we exploited the resistance of myeloma plasma cells to retroviral transduction by targeting the hematopoietic CD34+ cell population with a retroviral vector carrying a selectable marker (the truncated form of the human receptor for nerve growth factor, ΔNGFR). We performed therefore a further myeloma purging step by selecting the transduced cells at the end of the culture. Results Overall recovery of CD34+ cells after culture was 128.5%; ΔNGFR transduction rate was 28.8% for CD34+ cells and 0% for CD138-selected primary myeloma cells, respectively. Recovery of CD34+ cells after ΔNGFR selection was 22.3%. By patient-specific Ig-gene rearrangements, we assessed a decrease of 0.7–1.4 logs in tumor load after the CD34+ cell selection, and up to 2.3 logs after culture and ΔNGFR selection. Conclusion We conclude that ex-vivo culture and retroviral-mediated transduction of myeloma leukaphereses provide an efficient tumor cell purging.
Collapse
Affiliation(s)
- Sara Deola
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute H.S. Raffaele, Milan, Italy
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samantha Scaramuzza
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute H.S. Raffaele, Milan, Italy
| | - Roberto Sciarretta Birolo
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute H.S. Raffaele, Milan, Italy
| | | | - Francesca Ficara
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute H.S. Raffaele, Milan, Italy
| | - Jonathan Dando
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute H.S. Raffaele, Milan, Italy
| | - Claudia Voena
- Hematology and BMT Unit, Scientific Institute H.S. Raffaele, Milan, Italy
- Department of Biomedical Sciences and Human Oncology and Center for Experimental Research and Clinical Studies, University of Turin, Turin, Italy
| | - Sergio Vai
- Hematology and BMT Unit, Scientific Institute H.S. Raffaele, Milan, Italy
| | - Marta Monari
- Hematology and BMT Unit, Scientific Institute H.S. Raffaele, Milan, Italy
| | - Enrico Pogliani
- Hematology and Bone Marrow Transplant Unit, Hospital S. Gerardo, Monza, Italy
| | - Gianmarco Corneo
- Hematology and Bone Marrow Transplant Unit, Hospital S. Gerardo, Monza, Italy
| | - Jacopo Peccatori
- Hematology and BMT Unit, Scientific Institute H.S. Raffaele, Milan, Italy
| | - Silvia Selleri
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Human Morphology, University of Milan, Milan, Italy
| | - Claudio Bordignon
- Hematology and BMT Unit, Scientific Institute H.S. Raffaele, Milan, Italy
| | - Maria Grazia Roncarolo
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute H.S. Raffaele, Milan, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), Scientific Institute H.S. Raffaele, Milan, Italy
| | - Marco Bregni
- Hematology and BMT Unit, Scientific Institute H.S. Raffaele, Milan, Italy
| |
Collapse
|
19
|
Abstract
Gene-marking studies were the first gene-transfer protocols approved for human use. Their intent was not directly therapeutic but rather to track the behavior and fate of cells in vivo, and to use this information to improve treatment protocols. For more than fifteen years, gene-marking studies using retroviral vectors have provided invaluable information about the biology of human hematopoietic cells and T lymphocytes, and have helped guide cell therapies intended to treat malignant disease. Although the safety record of marking studies has been impeccable, the development of leukemia by immunodeficient children treated with retroviral vectors cast a pall over the entire field and essentially brought the era of pure gene-marking studies to an abrupt end. Paradoxically, the impetus these events gave to studying retroviral integration sites in host cell DNA emphasized the additional information that marker studies could provide about the behavior of cells at the clonal level. As confidence has slowly returned, marker studies have reappeared, usually as components of gene therapy protocols in which a marker gene or sequence is incorporated to allow the modified cells to be tracked or imaged in vivo. Hence, gene marking continues to have much to offer in terms of our understanding of the behavior, fate, and safety of gene-modified cells in vivo.
Collapse
Affiliation(s)
- Siok-Keen Tey
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas 77030, USA.
| | | |
Collapse
|
20
|
Trobridge G, Beard BC, Kiem HP. Hematopoietic stem cell transduction and amplification in large animal models. Hum Gene Ther 2006; 16:1355-66. [PMID: 16390267 DOI: 10.1089/hum.2005.16.1355] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Progress in retroviral gene transfer to large animal hematopoietic stem cells (HSCs) has led to efficient, reproducible long-term marking in both canine and nonhuman primate models. Successes for HSC gene therapy have occurred in the severe combined immunodeficiency setting, in which transduced cells have a selective advantage. However, for most diseases, the therapeutic transgene does not confer a sufficient survival advantage, and increasing the percentage of gene-marked cells in vivo will be necessary to observe a therapeutic effect. In vivo amplification should expand the potential of HSC gene therapy, and progress in this area has benefited greatly from the use of large animal models where efficacy and toxicity have often not correlated with results in murine models. To date, the best results have been observed with O(6)-methylguanine-DNA methyltransferase (MGMT) selection, with which increases in gene-marked repopulating cells have been maintained long-term, likely because of the toxicity of 1,3-bis-(2-chloroethyl)-1-nitrosourea and temozolomide to quiescent HSCs. Using MGMT selection, long-term marking levels exceeding 50% can now be routinely attained with minimal toxicity. There is cause to be optimistic that HSC gene therapy with in vivo amplification will soon allow the treatment of several genetic and infectious diseases.
Collapse
Affiliation(s)
- Grant Trobridge
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | |
Collapse
|
21
|
KELLY PATRICKF, CARRINGTON JODY, NATHWANI AMIT, VANIN ELIOF. RD114-Pseudotyped Oncoretroviral Vectors. Ann N Y Acad Sci 2006. [DOI: 10.1111/j.1749-6632.2001.tb03596.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Abstract
The lymphoproliferative disorders represent a large group of diseases with a significant variation in presentation and clinical course. There has been a trend of increasing incidence for some of these disorders, and despite advances in therapies, a significant number of patients either respond poorly or have early relapses. For this reason there is a need to investigate novel therapies to be used either alone or as adjunct treatment in combination with conventional therapies. Gene therapy is a relatively new field that takes advantage of our increased understanding of molecular biology with the aim of treating a variety of diseases including cancer. It is defined as the introduction of genetic material into cells for therapeutic intent. Methods to improve gene delivery efficiency have been the focus of a large amount of research and to date the optimal procedure uses viruses such as oncoretroviruses, lentiviruses, adenoviruses, adeno-associated viruses and herpes simplex viruses. There are four main gene therapy strategies that might be used for the treatment of lymphoproliferative disorders. First, immunotherapy using tumour vaccines or techniques to enhance the function of immune effector cells has been investigated with some success in patients with B-cell malignancies. Second, the introduction of prodrug-activated 'suicide' genes into cells has been explored, in particular in patients with post-transplantation lymphoproliferative disease. Third, direct lysis of tumour cells using viruses shows some early promise, especially in the treatment of B-cell disorders by manipulating the measles virus to target the CD20 antigen. Finally, anti-gene strategies such as anti-sense therapy, ribozymes, and most recently RNA interference, could be used to suppress expression of specific target genes. RNA interference in particular has tremendous potential and has been studied in the context of anaplastic large cell lymphoma as well as Epstein-Barr virus-associated malignancies. Whilst we are still in the early days of this field and to date results have been modest, there is still a significant potential for gene therapy to play a role in the future treatment of these disorders.
Collapse
Affiliation(s)
- Stephen R Larsen
- Gene and Stem Cell Therapy Program, Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Australia
| | | |
Collapse
|
23
|
Trobridge G, Beard BC, Kiem HP. Hematopoietic Stem Cell Transduction and Amplification in Large Animal Models. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.16.ft-144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
24
|
Ferguson C, Larochelle A, Dunbar CE. Hematopoietic stem cell gene therapy: dead or alive? Trends Biotechnol 2005; 23:589-97. [PMID: 16216357 DOI: 10.1016/j.tibtech.2005.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Revised: 06/28/2005] [Accepted: 09/23/2005] [Indexed: 01/21/2023]
Abstract
Despite some reports of toxicity in recent clinical trials, many scientists believe that the use of gene therapy in the treatment of congenital genetic defects and acquired disorders has too much potential to abandon. Hematopoietic stem cells (HSCs) have been primary targets for gene therapy owing to their capacity for differentiation and self-renewal, whereby multiple cell lineages can potentially be corrected for the lifetime of an individual. These efforts represent a long-term investment towards broadening physicians' treatment options for patients whose diseases, in particular certain immunodeficiencies, are fatal and where no other therapy is available. We review the recent progress and clinical triumphs as well as the reported toxicity related to insertional mutagenesis. We also discuss the current risk-to-benefit estimates and future strategies to reduce the risks and allow full realization of clinical potential. Scientists are continually revising protocols: going both from "bench to bedside" and, as strikingly demonstrated by HSC gene therapy, from "bedside to bench."
Collapse
Affiliation(s)
- Cole Ferguson
- Molecular Hematopoiesis Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
25
|
Glimm H, Schmidt M, Fischer M, Schwarzwaelder K, Wissler M, Klingenberg S, Prinz C, Waller CF, Lange W, Eaves CJ, von Kalle C. Efficient marking of human cells with rapid but transient repopulating activity in autografted recipients. Blood 2005; 106:893-8. [PMID: 15845903 PMCID: PMC1895162 DOI: 10.1182/blood-2004-07-2859] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Short-term hematopoietic reconstituting cells have been identified in mice, nonhuman primates, and among human cells that engraft xenogeneic hosts. We now present clonal marking data demonstrating a rapid but unsustained contribution of cultured human autografts to the initial phase of hematologic recovery in myeloablated patients. Three patients received transplants of granulocyte colony-stimulating factor-mobilized autologous peripheral blood (PB) cells, of which a portion (8%-25% of the CD34+ cells) had been incubated in vitro with growth factors (5 days) and clinical grade LN retrovirus (3-5 days). More than 9% of the clonogenic and long-term culture-initiating cells harvested were transduced. Semiquantitative and linear amplification-mediated polymerase chain reaction analyses of serial PB samples showed that marked white blood cells appeared in all 3 patients within 11 days and transiently constituted up to 0.1% to 1% of those produced in the first month. However, within another 2 to 9 months, marked cells had permanently decreased to very low levels. Analysis of more than 50 vector insertion sites showed none of the clones detected in the first month were active later. Eighty percent of inserts were located within or near genes, 2 near CXCR4. These findings provide direct evidence of cells with rapid but transient repopulating activity in patients and demonstrate their efficient transduction in vitro.
Collapse
Affiliation(s)
- Hanno Glimm
- Department of Internal Medicine I, Albert-Ludwigs-University, Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bollard CM, Aguilar L, Straathof KC, Gahn B, Huls MH, Rousseau A, Sixbey J, Gresik MV, Carrum G, Hudson M, Dilloo D, Gee A, Brenner MK, Rooney CM, Heslop HE. Cytotoxic T lymphocyte therapy for Epstein-Barr virus+ Hodgkin's disease. ACTA ACUST UNITED AC 2005; 200:1623-33. [PMID: 15611290 PMCID: PMC2211993 DOI: 10.1084/jem.20040890] [Citation(s) in RCA: 294] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Epstein Barr virus (EBV)+ Hodgkin's disease (HD) expresses clearly identified tumor antigens derived from the virus and could, in principle, be a target for adoptive immunotherapy with viral antigen–specific T cells. However, like most tumor-associated antigens in immunocompetent hosts, these potential targets are only weakly immunogenic, consisting primarily of the latent membrane protein (LMP)1 and LMP2 antigens. Moreover, Hodgkin tumors possess a range of tumor evasion strategies. Therefore, the likely value of immunotherapy with EBV-specific cytotoxic effector cells has been questioned. We have now used a combination of gene marking, tetramer, and functional analyses to track the fate and assess the activity of EBV cytotoxic T lymphocyte (CTL) lines administered to 14 patients treated for relapsed EBV+ HD. Gene marking studies showed that infused effector cells could further expand by several logs in vivo, contribute to the memory pool (persisting up to 12 mo), and traffic to tumor sites. Tetramer and functional analyses showed that T cells reactive with the tumor-associated antigen LMP2 were present in the infused lines, expanded in peripheral blood after infusion, and also entered tumor. Viral load decreased, demonstrating the biologic activity of the infused CTLs. Clinically, EBV CTLs were well tolerated, could control type B symptoms (fever, night sweats, and weight loss), and had antitumor activity. After CTL infusion, five patients were in complete remission at up to 40 mo, two of whom had clearly measurable tumor at the time of treatment. One additional patient had a partial response, and five had stable disease. The performance and fate of these human tumor antigen–specific T cells in vivo suggests that they might be of value for the treatment of EBV+ Hodgkin lymphoma.
Collapse
Affiliation(s)
- Catherine M Bollard
- Center for Gene and Cell Therapy, Baylor College of Medicine, 6621 Fannin St., MC 3-3320, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Alajez NM, Schmielau J, Alter MD, Cascio M, Finn OJ. Therapeutic potential of a tumor-specific, MHC-unrestricted T-cell receptor expressed on effector cells of the innate and the adaptive immune system through bone marrow transduction and immune reconstitution. Blood 2005; 105:4583-9. [PMID: 15746083 PMCID: PMC1894994 DOI: 10.1182/blood-2004-10-3848] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
T-cell receptor (TCR) with unique major histocompatibility complex (MHC)-unrestricted antigen-binding properties was isolated from a human T-cell clone specific for the tumor antigen MUC1. This TCR binds its epitope on the MUC1 protein without the requirement of processing and presentation. A single-chain Valpha/Vbeta/Cbeta (scTCR) was fused to a CD3 zeta (zeta) chain to allow expression on the surface of cells of the innate (granulocytes, macrophages, natural killer [NK] cells) as well as the adaptive (T and B cells) immune system. To test the ability of the cells of the innate immune system to reject a tumor when provided with a tumor antigen-specific TCR, we reconstituted severe combined immunodeficiency (SCID) mice with bone marrow cells transduced with a retroviral vector encoding this receptor and challenged them with a MUC1-positive human tumor. These mice controlled the growth of the tumor significantly better than the control mice. We performed a similar experiment in immunocompetent mice transgenic for human MUC1. Expression of the TCR on large percentages of cells did not result in infiltration or destruction of tissues expressing MUC1. Reconstituted mice controlled the outgrowth of a MUC1-transfected but not the parental control tumor. scTCR expression appears lifelong, suggesting a successful transduction of the self-renewing stem cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/chemistry
- B-Lymphocytes/immunology
- Bone Marrow Cells/cytology
- CD3 Complex/chemistry
- Cell Line, Tumor
- Cell Lineage
- Enzyme-Linked Immunosorbent Assay
- Epitopes/chemistry
- Flow Cytometry
- Granulocytes/cytology
- Green Fluorescent Proteins/metabolism
- Hematopoietic Stem Cells/cytology
- Humans
- Immune System
- Immunohistochemistry
- Immunotherapy/methods
- Interleukin-2/metabolism
- Killer Cells, Natural/cytology
- Macrophages/cytology
- Major Histocompatibility Complex
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, SCID
- Mice, Transgenic
- Microscopy, Fluorescence
- Molecular Sequence Data
- Mucin-1/chemistry
- Neoplasm Transplantation
- Neoplasms/therapy
- Rats
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/therapeutic use
- Retroviridae/genetics
- Sequence Homology, Amino Acid
- Software
- Stem Cells/cytology
- T-Lymphocytes/immunology
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Nehad M Alajez
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Gene therapy aims to correct the disease process by restoring, modifying or enhancing cellular functions through the introduction of a functional gene into a target cell. Whilst the concept of gene therapy is simple, the practical reality of translating this new technology to the clinic has proven to be more difficult than first imagined. Recent progress in gene transfer technology has shown impressive clinical success in infants with immunodeficiency. However, two of these children have subsequently developed leukaemia as a result of insertional mutagenesis, thus, raising important questions about the safety of genetic therapeutics. This article reviews the current status of gene therapy and outlines the challenges faced by this emerging technology that holds so much promise for many suffering from catastrophic disorders.
Collapse
Affiliation(s)
- Amit C Nathwani
- Department of Haematology, University College London, London, UK.
| | | | | |
Collapse
|
29
|
Abstract
Over the past two decades, the ability to transfer genes into hematopoietic stem cells (HSCs) has provided new insights into the behavior of individual stem cells and offered a novel approach for the treatment of various inherited or acquired disorders. At present, gene transfer into HSCs has been achieved mainly using modified retroviruses. While retrovirus-based vectors could efficiently transduce murine HSCs, extrapolation of these methods to large mammals and human clinical trials resulted in very low numbers of gene-marked engrafted cells. In addition, in vitro progenitor assays used to optimize gene transfer procedures were found to poorly predict the outcome of stem cell gene transfer. The focus rapidly turned to the development of superior and more relevant preclinical assays in human stem cell gene transfer research. Xenogeneic transplant models and large animal transplantation system have been invaluable. The development of better assays for evaluating human gene therapy protocols and a better understanding of stem cell and vector biology has culminated over the past decade in multiple strategies to improve gene transfer efficiency into HSCs. Improved gene transfer vectors, optimization of cytokine combination, and incorporation of a recombinant fragment of fibronectin during transduction are examples of novel successful additions to the early gene transfer protocols that have contributed to the first unequivocal clinical benefits resulting from genetic manipulation of HSC.
Collapse
Affiliation(s)
- André Larochelle
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
30
|
Zielske SP, Braun SE. Cytokines: Value-Added Products in Hematopoietic Stem Cell Gene Therapy. Mol Ther 2004; 10:211-9. [PMID: 15294167 DOI: 10.1016/j.ymthe.2004.05.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2004] [Accepted: 05/17/2004] [Indexed: 10/26/2022] Open
Affiliation(s)
- Steven P Zielske
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA 01605, USA.
| | | |
Collapse
|
31
|
Jabbour E, Keating MJ, Champlin RE, Khouri IF. Stem cell transplantation for chronic lymphocytic leukemia: should not more patients get a transplant? Bone Marrow Transplant 2004; 34:289-97. [PMID: 15220957 DOI: 10.1038/sj.bmt.1704593] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Novel therapeutic approaches with conventional chemotherapy and monoclonal antibody combinations have improved the complete remission rates in chronic lymphocytic leukemia. However, cure remains elusive, particularly in fludarabine-refractory patients, whose prognosis remains poor. Autologous stem cell transplantation (SCT) has been explored for such patients, lengthening the time to treatment failure in selected patients, but there is little hope that it will improve the cure rate. The strategy is particularly ineffective in patients with poor biological prognostic factors, such as abnormal cytogenetics and unmutated immunoglobulin heavy-chain variable region. Allogeneic SCT remains the only curative approach, producing an extended disease-free survival in 25-60%, mainly via the graft-versus-leukemia effect. The treatment-related mortality with such an approach has been significant, however, with a 30-40% risk of death within 100 days of the transplant. Nonmyeloablative (NMA) conditioning regimens may produce high response rates and lower morbidity, especially for patients with chemosensitive disease. Randomized trials designed according to the new biologic prognostic parameters described in chronic lymphocytic leukemia are required to better define the role of NMA SCT in the near future.
Collapse
Affiliation(s)
- E Jabbour
- Department of Blood and Bone Marrow Transplant, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
32
|
Abstract
The cure rate for cancer in children is currently almost 75%. This rate has remained fairly constant over the past few years, which suggests that the limits of today's curative treatment potential have been reached. The development of cell therapy techniques opens up new therapeutic possibilities in paediatric oncology. Here, we deal both with a number of cell therapy techniques, which have already proved their efficacy in children, and other more innovative approaches, which require validation. Examples of the use of autologous and allogeneic cells are described. Clinical studies and their results, while often preliminary, are reported. The importance of well run clinical research, a clear and progressive legal framework and the necessary substantial economic support for the development of cell therapy are underlined.
Collapse
Affiliation(s)
- J Kanold
- Unité bioclinique de thérapie cellulaire, service de pédiatrie B, Hôtel-Dieu, C.H.U., B.P.69, 11, boulevard Léon Malfreyt, 63003 Clermont-Ferrand, France.
| | | | | | | |
Collapse
|
33
|
Affiliation(s)
- M K Brenner
- Center for Gene and Cell Therapy, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
34
|
Affiliation(s)
- E M Horwitz
- Division of Stem Cell Transplantation, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | |
Collapse
|
35
|
Abstract
Despite advances in the treatment of neuroblastoma (NBL), recurrence and metastases continue to pose major problems in clinical management. The relation between micrometastases and the development of secondary disease is not fully understood. However, accurate methods to detect low numbers of tumour cells may allow the evaluation of their role in the disease process, and by implication the possible benefits of eliminating them. Although there is substantial evidence for the increased sensitivity of current molecular methods for the detection of NBL cells compared with more conventional cytology, the clinical relevance and usefulness of detecting this disease remain controversial. The primary goal of current translational research must be to evaluate the clinical relevance of micrometastatic disease detected by these methods in multicentre prospective clinical outcome studies. Only then can the clinical usefulness of these methods be defined so that they may be introduced into relevant clinical practice.
Collapse
Affiliation(s)
- S A Burchill
- Children's Cancer Research Laboratory, Cancer Research UK Clinical Centre, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK.
| |
Collapse
|
36
|
Bhatia M. The ultimate source of human hematopoietic stem cells: thinking outside the marrow. CLONING AND STEM CELLS 2003; 5:89-97. [PMID: 12713705 DOI: 10.1089/153623003321512201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The ability to reconstitute cellular components of the hematopoietic system has immense utility in several areas of clinical medicine. These include replacement of cells responsible for innate and acquired immunity, providing red cells for oxygen transport, and ultimately the ability to recover hematopoietic function by repopulating all lineages comprising the entire blood system. This latter property functionally defines the mammalian hematopoietic stem cell (HSC). Recently, human embryonic stem cells (ESCs) have been suggested to be a viable source of transplantable hematopoietic cells. Although the number of human ESCs is virtually unlimited, the ability to efficiently differentiate adequate numbers of cells that possess hematopoietic repopulating ability remains to be determined. Achieving this goal is confounded by the difficulty of experimentally generating murine hematopoietic cell types capable of in vivo reconstitution from mouse ESC, suggesting that similar limitations may arise using human counterparts. Although the use of human ESCs and adult somatic HSCs have their independent merits, a direct comparison between HSCs derived from each source using similar assays will ultimately be required to determine the best source for clinical use. Here we will summarize the results from efforts to differentiate and assay primitive hematopoietic cells derived from ESCs, and compare these findings to similar parameters using putative mammalian HSCs harvested from the adult.
Collapse
Affiliation(s)
- Mickie Bhatia
- Robarts Research Institute, Stem Cell Biology and Regenerative Medicine, London, Ontario, Canada.
| |
Collapse
|
37
|
Bueren JA, Guenechea G, Casado JA, Lamana ML, Segovia JC. Genetic modification of hematopoietic stem cells: recent advances in the gene therapy of inherited diseases. Arch Med Res 2003; 34:589-99. [PMID: 14734099 DOI: 10.1016/j.arcmed.2003.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hematopoietic stem cells constitute a rare population of precursor cells with remarkable properties for being used as targets in gene therapy protocols. The last years have been particularly productive both in the fields of gene therapy and stem cell biology. Results from ongoing clinical trials have shown the first unquestionable clinical benefits of immunodeficient patients transplanted with genetically modified autologous stem cells. On the other hand, severe side effects in a few patients treated with gene therapy have also been reported, indicating the usefulness of further improving the vectors currently used in gene therapy clinical trials. In the field of stem cell biology, evidence showing the plastic potential of adult hematopoietic stem cells and data indicating the multipotency of adult mesenchymal precursor cells have been presented. Also, the generation of embryonic stem cells by means of nuclear transfer techniques has appeared as a new methodology with direct implications in gene therapy.
Collapse
Affiliation(s)
- Juan A Bueren
- Hematopoietic Gene Therapy Program, Comisión Interministerial de Ciencia y Tecnología/Fundación Marcelino Botín, Madrid, Spain.
| | | | | | | | | |
Collapse
|
38
|
Joyce RM, Regan M, Ottaway J, Umiel T, Tetreault JC, Levine J, McDermott D, Hurley D, Giallombardo N, Smith T, Lamontagne D, Uhl L, Avigan D. A phase I-II study of rituximab, ifosfamide, mitoxantrone and etoposide (R-IME) for B cell non-Hodgkin's lymphoma prior to and after high-dose chemotherapy and autologous stem cell transplantation (HDC-ASCT). Ann Oncol 2003; 14 Suppl 1:i21-7. [PMID: 12736227 DOI: 10.1093/annonc/mdg705] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
This phase I-II study describes the safety of rituximab, ifosfamide, mitoxantrone and etoposide (R-IME) as an induction regimen prior to high-dose chemotherapy and autologous stem cell transplantation (HDC-ASCT), and rituximab given post-HDC-ASCT for B cell non-Hodgkins's lymphoma. This study also measured the effect on disease burden and stem cell contamination. Patients with relapsed, refractory or poor risk B cell lymphomas were eligible. Patients were treated with two cycles of R-IME; all non-progressing patients under-went a third cycle and peripheral blood stem cell (PBSC) collection. Patients underwent HDC-ASCT and those patients in remission after HDC-ASCT were treated with four additional doses of rituximab. Tumor cell contamination was measured at baseline and in the PBSC. Serial immunoglobulin levels were measured. Patients were followed for time to treatment failure (TTF) and overall survival (OS). Thirty-two patients were enrolled. Thirty patients had at least stable disease after two cycles of R-IME. Twenty-nine underwent stem cell collection. The response rate to R-IME induction was 77% (20/26) with 35% (9/26) complete response(CR). Stem cell mobilization was successful in 93% (27/29) of patients. The response rate to R-IME induction and HDC-ASCT was 95% with a confirmed CR of 68%. Median follow-up was 28 months; the median TTFand OS have not been reached. There was a significant decline in stem cell tumor cell contamination and a significant decline in IgG without an increase in infections. Forty-three per cent of patients had transient neutropenia after post-transplant rituximab. R-IME is an effective cytoreductive and mobilization regimen. There appears to be a reduction in the number of lymphoma cells in the stem cell product and the toxicity is manageable.
Collapse
Affiliation(s)
- R M Joyce
- Harvard Medical School, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Garcia-Castro J, Rio P, Lillo R, Garcia-Sanchez F, Segovia JC, Bueren JA. Purging of leukemia-contaminated bone marrow grafts using suicide adenoviral vectors: an in vivo murine experimental model. Gene Ther 2003; 10:1328-35. [PMID: 12883529 DOI: 10.1038/sj.gt.3301993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Autologous bone marrow transplantation is an alternative therapeutic option for acute myeloid leukemia patients lacking a compatible donor. However, bone marrow from these patients may contain residual leukemic cells that should be ideally eliminated prior to the infusion of the graft. With the aim of developing more efficient protocols of graft purging, adenoviral-mediated gene transfer protocols have been conducted. We studied whether suicide adenoviral vectors expressing the cytosine deaminase gene (AdCD) could be used for selectively killing leukemic WEHI-3B cells. The AdCD transduction followed by the 5-fluorocytosine exposure abrogated the growth of WEHI-3B cells in vitro, with a minimal effect on normal hematopietic progenitors. To test the efficacy of the purging protocol in vivo, bone marrow cells were mixed with syngenic WEHI-3B cells and this chimeric cell population was transduced with AdCD vectors. Infected cells were injected into myeloablated Balb-c mice, which then received a 5-fluorocytosine treatment for 4 days. All mice transplanted with unpurged bone marrow developed leukemia and died. However, 90% of recipients receiving the purging treatment were healthy up to 9 months post-transplantation and had a perfectly re-established hematopoietic system, without any signal of leukemic cell presence. In conclusion, suicide adenoviral vectors are proposed as a tool for the purging of Adenoviral-susceptible myeloid leukemia cells contaminating autologous bone marrow grafts.
Collapse
Affiliation(s)
- J Garcia-Castro
- Gene Therapy Program, CIEMAT/Fundación Marcelino Botín, Av. Complutense, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
40
|
Vollweiler JL, Zielske SP, Reese JS, Gerson SL. Hematopoietic stem cell gene therapy: progress toward therapeutic targets. Bone Marrow Transplant 2003; 32:1-7. [PMID: 12815471 DOI: 10.1038/sj.bmt.1704081] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The concept of hematopoietic stem cell gene therapy is as exciting as that of stem cell transplantation itself. The past 20 years of research have led to improved techniques for transferring and expressing genes in hematopoietic stem cells and preclinical models now routinely indicate the ease with which new genes can be expressed in repopulating stem cells of multiple species. Both modified murine oncoretroviruses and lentiviruses transmit genes into the genome of hematopoietic stem cells and allow expression in the host following transplantation. Using oncoretroviruses, therapeutic genes for severe combined immunodeficiency, common variable gamma chain immunodeficiency, chronic granulomatous disease, Hurler's and Gaucher's Disease have all been used clinically with only modest success except for the patients with immunodeficiency in whom a partial T-cell chimerism has been dramatic. Since stem cell selection in vivo appears important to the therapeutic success of gene transfer, drug resistance selection, most recently using the MGMT gene, has been developed and appears to be safe. Future trials combining a drug resistance and therapeutic gene are planned, as are trials using safety-modified lentiviruses. The therapeutic potential of hematopoietic stem cell gene therapy, particularly given recent advances in stem cell plasticity, remains an exceptionally exciting area of clinical research.
Collapse
Affiliation(s)
- J L Vollweiler
- Division of Hematology-Oncology, Comprehensive Cancer Center at University Hospitals of Cleveland, Case Western Reserve University School of Medicine, USA
| | | | | | | |
Collapse
|
41
|
Fruehauf S, Veldwijk MR, Berlinghoff S, Basara N, Baum C, Flasshove M, Hegewisch-Becker S, Kröger N, Licht T, Moritz T, Hengge UR, Zeller WJ, Laufs S. Gene therapy for sarcoma. Cells Tissues Organs 2003; 172:133-44. [PMID: 12426490 DOI: 10.1159/000065614] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Soft tissue sarcomas are mesenchymal tumors which respond poorly to systemic therapy. Recent studies suggest a higher response rate with an increased doxorubicin dosage. However, this was parallel with a profound hematotoxicity in 75% of patients. Transfer of the human multidrug resistance 1 (MDR1) gene to normal hematopoietic stem cells and transplantation may significantly reduce the hematotoxicity of anthracyclin-based chemotherapy. To test this concept of supportive gene therapy in advance of a clinical study, we transduced mobilized peripheral blood progenitor cells (PBPC) with the retroviral vector SF91m3 containing the human MDR1 gene, transplanted these cells to immune-deficient mice, allowed 6 weeks for engraftment to occur and treated the animals with MDR1-based chemotherapy. In the MDR1-transduced group the human leukocytes were significantly protected from the toxicity of chemotherapy (p < 0.05). While the gene transfer rate was in the range of 10% and thus comparable to recent clinical trials, the gene expression was 59% of transduced cells and thus significantly higher than previously reported for less-advanced vectors. On the other hand, ifosfamide, a drug which has been used successfully for stem cell mobilization, is active in soft tissue sarcoma. Due to these favorable characteristics sarcoma is an attractive target to test the efficacy of MDR1 gene therapy in a clinical setting. Gene therapeutic strategies may also be used to directly target sarcoma cells, e.g. by transfer of suicide genes. We found that adenoassociated virus 2 (AAV-2) vectors efficiently transduce human HS-1 and HT1080 sarcoma cells (>90%) while other tumor cell lines and primary human PBPC were less susceptible. The thymidine kinase (TK) suicide gene was cloned into an AAV-2 vector and a complete kill of TK-transduced HS-1 and HT1080 cells was observed following exposure to aciclovir or ganciclovir (GCV), while >90% of mock-transduced HS-1 cells survived at these dosages. Transplantation of those sarcoma cells to nonobese diabetic (NOD)/LtSz-severe-combined immunodeficient (scid)/scid (NOD/SCID) mice resulted in a survival of >5 months in the AAV-TK-transduced/GCV-treated group, while the mice in the mock-transduced/GCV-treated group had died after 3 weeks. These data show that soft tissue sarcomas are a particularly suitable model system for the development and clinical testing of new gene therapeutic concepts.
Collapse
Affiliation(s)
- S Fruehauf
- Department of Internal Medicine V, University of Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Krance RA, Kuehnle I, Rill DR, Mei Z, Pinetta C, Evans W, Brown MP, Pulé M, Heslop HE, Brenner MK. Hematopoietic and immunomodulatory effects of lytic CD45 monoclonal antibodies in patients with hematologic malignancy. Biol Blood Marrow Transplant 2003; 9:273-81. [PMID: 12720220 DOI: 10.1053/bbmt.2003.50024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The CD45 antigen is present on all cells of the hematopoietic lineage. In some rodent models, lytic CD45 monoclonal antibodies (MAbs) induce complete marrow aplasia. In others, only transient myelolymphodepletion are observed, which are nonetheless sufficient to permit engraftment with fully allogeneic stem cells after otherwise ineffective doses of radiation. The in vivo effects of unconjugated cytolytic CD45 MAbs on myeloid and lymphoid cells in humans are unknown, so it is unclear if they could contribute in a similar way to conventional ablative or to nonmyeloablative preparative regimens used for stem cell transplantation (SCT). We therefore assessed the safety, myeloreductive activities, and lymphoreductive activities of the unconjugated rat anti-human CD45 MAbs, YTH25.4 and YTH54.12, in subjects who were to undergo SCT for advanced hematologic malignancy. The MAb pair bind to contiguous but nonoverlapping epitopes on CD45 and work synergistically to fix complement and recruit cellular lytic mechanisms. The MAbs were given in increasing doses up to 1600 microg/kg during 4 days, after which the patients began their conventional transplantation preparative regimen. The maximum tolerated dose of these MAbs, 400 microg/kg/d, produced marked reduction in circulating lymphoid and myeloid cells while largely sparing marrow progenitors. In 2 of 3 patients who had active leukemia at the time of study, the MAbs reduced the percentage of leukemic blast cells in bone marrow. Seven of 14 patients are disease free 610 to 1555 days post-SCT. The in vivo myeloreductive and lymphoreductive properties of lytic CD45 MAb in humans, therefore, closely parallel the activity seen in a murine model and, therefore, may be of similar value.
Collapse
Affiliation(s)
- Robert A Krance
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Biagi E, Bollard C, Rousseau R, Brenner M. Gene Therapy for Pediatric Cancer: State of the Art and Future Perspectives. J Biomed Biotechnol 2003; 2003:13-24. [PMID: 12686719 PMCID: PMC179759 DOI: 10.1155/s1110724303209025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
While modern treatments have led to a dramatic improvement in survival for pediatric malignancy, toxicities are high and a significant proportion of patients remain resistant. Gene transfer offers the prospect of highly specific therapies for childhood cancer. "Corrective" genes may be transferred to overcome the genetic abnormalities present in the precancerous cell. Alternatively, genes can be introduced to render the malignant cell sensitive to therapeutic drugs. The tumor can also be attacked by decreasing its blood supply with genes that inhibit vascular growth. Another possible approach is to modify normal tissues with genes that make them more resistant to conventional drugs and/or radiation, thereby increasing the therapeutic index. Finally, it may be possible to attack the tumor indirectly by using genes that modify the behavior of the immune system, either by making the tumor more immunogenic, or by rendering host effector cells more efficient. Several gene therapy applications have already been reported for pediatric cancer patients in preliminary Phase 1 studies. Although no major clinical success has yet been achieved, improvements in gene delivery technologies and a better understanding of mechanisms of tumor progression and immune escape have opened new perspectives for the cure of pediatric cancer by combining gene therapy with standard therapeutic available treatments.
Collapse
Affiliation(s)
- Ettore Biagi
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Catherine Bollard
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Raphael Rousseau
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Malcolm Brenner
- Center for Cell and Gene Therapy, Department of Pediatrics-Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
44
|
Abstract
The use of high-dose chemotherapy followed by autologous HCT and the use of allogeneic HCT in children and adolescents with high-risk ALL, AML, and NBL has successfully improved outcomes. For other diseases, however, the role of HCT in treatment remains a subject of further research. The availability of HCT was significantly expanded by developing alternative graft sources that currently include BM, peripheral blood, and UCB from autologous and allogeneic related or unrelated donors. Progress in autologous HCT has been achieved by the identification of more effective and less toxic preparative regimens and by ex vivo purging of stem cell products. In allogeneic HCT, graft-versus-leukemia or graft-versus-tumor effects are being exploited increasingly to lower relapse rates. In addition, immunomodulation to promote tolerance, as well as allogeneic antitumor reactions have been achieved by antibody therapy, cytokine therapy, or cell-based immunotherapy. Future improvements are likely, as evidenced by promising preliminary results in the development of stem cell collection techniques, in vitro stem cell expansion, and purging techniques of stem cell grafts. The development of less intensive or nonmyeloablative preparative regimens may further reduce regimen-related morbidity and mortality Specific immunotherapy may facilitate tolerance induction in mismatched allogeneic HCT and support allogeneic HCT in the setting of donor-host HLA disparity. Ultimately, advances in cytokine therapy, tumor-specific vaccines, and gene therapy may decrease or even eradicate recurrence of the malignant disease after HCT.
Collapse
Affiliation(s)
- Ulrike Reiss
- Department of Hematoloy/Oncology, Children's Hospital and Research Center at Oakland, 747 52nd Street Oakland, CA 94609, USA.
| | | |
Collapse
|
45
|
Nicolini FE, Imren S, Oh IH, Humphries RK, Leboulch P, Fabry ME, Nagel RL, Eaves CJ. Expression of a human beta-globin transgene in erythroid cells derived from retrovirally transduced transplantable human fetal liver and cord blood cells. Blood 2002; 100:1257-64. [PMID: 12149206 DOI: 10.1182/blood-2002-02-0599] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transfer of therapeutic genes to human hematopoietic stem cells (HSCs) using complex vectors at clinically relevant efficiencies remains a major challenge. Recently we described a stable retroviral vector that sustains long-term expression of green fluorescent protein (GFP) and a human beta-globin gene in the erythroid progeny of transduced murine HSCs. We now report the efficient transduction of primitive human CD34(+) fetal liver or cord blood cells with this vector and expression of the beta-globin transgene in the erythroid progeny of these human cells for at least 2 months. After growth factor prestimulation and then a 2- to 3-day exposure to the virus, 35% to 55% GFP(+) progeny were seen in assays of transduced colony-forming cells, primitive erythroid precursors that generate large numbers of glycophorin A(+) cells in 3-week suspension cultures, and 6-week long-term culture-initiating cells. In immunodeficient mice injected with unselected infected cells, 5% to 15% of the human cells regenerated in the marrow (including the erythroid cells) were GFP(+) 3 and 6 weeks after transplantation. Importantly, the numbers of GFP(+) human lymphoid and either granulopoietic or erythroid cells in individual mice 6 weeks after transplantation were significantly correlated, indicative of the initial transduction of human multipotent cells with in vivo repopulating activity. Expression of the transduced beta-globin gene in human cells obtained directly from the mice or after their differentiation into erythroid cells in vitro was demonstrated by reverse transcriptase-polymerase chain reaction using specific primers. These experiments represent a significant step toward the realization of a gene therapy approach for human beta-globin gene disorders.
Collapse
Affiliation(s)
- Franck E Nicolini
- Terry Fox Laboratory, British Columbia Cancer Agency and University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Hess DA, Levac KD, Karanu FN, Rosu-Myles M, White MJ, Gallacher L, Murdoch B, Keeney M, Ottowski P, Foley R, Chin-Yee I, Bhatia M. Functional analysis of human hematopoietic repopulating cells mobilized with granulocyte colony-stimulating factor alone versus granulocyte colony-stimulating factor in combination with stem cell factor. Blood 2002; 100:869-78. [PMID: 12130497 DOI: 10.1182/blood.v100.3.869] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Using in vitro progenitor assays, serum-free in vitro cultures, and the nonobese diabetic/severe combined immune-deficient (NOD/SCID) ecotropic murine virus knockout xenotransplantation model to detect human SCID repopulating cells (SRCs) with multilineage reconstituting function, we have characterized and compared purified subpopulations harvested from the peripheral blood (PB) of patients receiving granulocyte colony-stimulating factor (G-CSF) alone or in combination with stem cell factor (SCF). Mobilized G-CSF plus SCF PB showed a 2-fold increase in total mononuclear cell content and a 5-fold increase in CD34-expressing cells depleted for lineage-marker expression (CD34(+)Lin(-)) as compared with patients treated with G-CSF alone. Functionally, G-CSF plus SCF-mobilized CD34(+)CD38(-)Lin(-) cells contained a 2-fold enhancement in progenitor frequency as compared with G-CSF-mobilized subsets. Despite enhanced cellularity and progenitor capacity, G-CSF plus SCF mobilization did not increase the frequency of SRCs as determined by limiting dilution analysis by means of unfractionated PB cells. Purification of SRCs from these sources demonstrated that as few as 1000 CD34(+)CD38(-)Lin(-) cells from G-CSF-mobilized PB contained SRC capacity while G-CSF plus SCF-mobilized CD34(+)CD38(-)Lin(-) cells failed to repopulate at doses up to 500 000 cells. In addition, primitive CD34(-)CD38(-)AC133(+)Lin(-) cells derived from G-CSF plus SCF-mobilized PB were capable of differentiation into CD34-expressing cells, while the identical subfractions from G-CSF PB were unable to produce CD34(+) cells in serum-free cultures. Our study defines qualitative and quantitative distinctions among subsets of primitive cells mobilized by means of G-CSF plus SCF versus G-CSF alone, and therefore has implications for the utility of purified repopulating cells from these sources.
Collapse
Affiliation(s)
- David A Hess
- Developmental Stem Cell Biology and Regenerative Medicine, John P. Robarts Research Institute, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
There has been important progress in the treatment of Acute Myeloid Leukaemia (AML) in patients under 60 years. A remission rate of 80% can be achieved by several schedules, and 40-45% of patients diagnosed will survive. It may still be possible to improve remission induction treatment eg by intensifying the Ara-C dose although may this only be detectable in an improved disease free survival. The is to reduce relapse. The risk main challenge is pre-determined by a number of powerful risk factors. In the experience of the MRC age, cytogenetics and clearance of blasts from the bone marrow after course 1. Using the later two in combination good risk patients (FAB M3, t(8;21) t(15;17) inv(16)) have a relapse risk of 32%. Poor risk (blasts >15% after course 1 or abnormalities of Chs 5 or 7, 3q- and complex changes have a relapse risk of 82%. All other cases are standard risk and ve a relapse risk of 56%. FLT3 mutations have been detected in about 25% of cases and provide additional negative predictive value overall and within each risk group. The assessment of the most effective consolidation treatment must be made taking into account the heterogeneity of the relapse risk. The MRC investigated the role of allo and autoBMT in addition to intensive chemotherapy. The data was analysis on an intent-to-treat or donor vs no donor basis. Although both types of transplant were able to reduce relapse overall and in all risk groups, there was an overall survival advantage only in standard risk patients. Since chemotherapy has improved since this study, there remains uncertainty about the benefit of transplant in all risk groups. Overall this experience has demonstrated that relapse can be reduced with more therapy. It is probable that the limits of conventional chemotherapy have been reached. The new AML15 trial will assess the value of adding the immunoconjugate (Mylotarg) to induction and/or chemotherapy. Improvements in older patients have been less detectable. MRC trials over the last 20 years show an improvement in remission rate (now 65%) but persistent poor survival (12% at 5 years). In the MRC AML11 Trial three induction schedules were compared (DAT vs ADE vs MAC) with DAT being superior. A comparison of a total of 3 vs 6 courses of treatment or the addition of interferon maintenance did not improve results. Newer approaches currently being assessed include resistance modulation; addition of immunoconjugate and minigrafts. New targets for treatment are emerging of which the most interesting is FLT3 inhibitors.
Collapse
Affiliation(s)
- Adrian Newland
- Department of Haematology, Barts and the London School of Medicine and Dentistry, UK
| |
Collapse
|
48
|
Hájek R, Korístek Z, Vinklárková J, Janovská E, Klabusay M, Doubek M, Dvoráková D, Bourková L, Dusek L, Büchler T, Adler J, Adam Z, Penka M, Mayer J, Vorlícek J. Interleukin-2 activation of haematopoietic stem cells. ACTA MEDICA AUSTRIACA 2002; 29:61-7. [PMID: 12050948 DOI: 10.1046/j.1563-2571.2002.02005.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Recent findings concerning the role of immunity in the eradication of residual malignant disease after autologous haematopoietic stem cell transplantation have led to extensive studies of T-cell and natural killer (NK) mediated anti-tumour effects. Interleukin 2 (IL-2) activation of autologous bone marrow (BM) or peripheral blood stem cells (PBSC) before transplantation is one of the methods of adoptive cell therapy. METHODS Autologous BM of patients with chronic myelogenous leukaemia (n = 11) and PBSC of patients with multiple myeloma (n = 14) were activated by IL-2 in laboratory conditions with the aim of evaluating the feasibility of this method, the activation of T and NK cells, recovery of active progenitor cells, microbial contamination, and reduction of malignant cell content. RESULTS Samples of BM (mean 2.6 x 10(6) cells) and PBSC (mean 10.3 x 10(6) cells) were cultured in complete culture medium with IL-2 (6000 Ul/ml) for 24 h. The recovery of CD34+ cells and CFU-GM was 82.5% and 51.5%, respectively, for BM, and 85% and 86%, respectively, for PBSC (mean values). No purging effect was detected by flow cytometry and a small decline in malignant cell contamination was observed by quantitative PCR in BM samples. No microbial contamination occurred during the sample processing. CONCLUSIONS The described in vitro activation of BM and peripheral blood stem cells using IL-2 was evaluated as a safe and reliable method suitable for clinical application.
Collapse
Affiliation(s)
- R Hájek
- Department of Internal Medicine-Hematooncology, Department of Clinical Hematology, University Hospital Brno, Bohunice, Jihlavská 20, CZ-639 00 Brno, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Moritz T, Williams DA. Marrow protection - transduction of hematopoietic cells with drug resistance genes. Cytotherapy 2002; 3:67-84. [PMID: 12028830 DOI: 10.1080/14653240152584640] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- T Moritz
- Department of Internal Medicine (Cancer Research), West German Tumor Center, University of Essen Medical School, Essen, Germany
| | | |
Collapse
|
50
|
Nakamura T, Sato K, Hamada H. Effective gene transfer to human melanomas via integrin-targeted adenoviral vectors. Hum Gene Ther 2002; 13:613-26. [PMID: 11916485 DOI: 10.1089/10430340252837215] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The utility of recombinant adenoviral vectors (Adv) for gene therapy is limited by their low transduction efficiency and lack of specificity for target cells. The low transduction efficiency is often recognized as due to deficiency of the primary adenoviral receptor, the coxsackievirus-adenovirus receptor (CAR). In this paper, studies of CAR levels on human melanoma cell lines confirmed that low transduction efficiency was closely related to deficiency of the adenoviral receptor. To achieve CAR-independent gene transfer via Adv, we modified viral tropism via genetic alteration of the adenovirus type 5 (Ad5) fiber protein. Insertion of an Arg-Gly-Asp (RGD)-containing peptide in the HI loop of the fiber knob domain allowed the virus to use an alternative receptor, the integrin receptor, during the cell entry process. With this modified vector (Adv-F/RGD) transduction was increased 5- to 96-fold relative to a vector containing wild-type fiber (Adv-F/wt) in five human melanoma cells expressing integrins of the alpha(v)beta(3), alpha(v)beta(5) class, which are recognized by the RGD peptide motif. In contrast, no significant difference in transduction efficiency between Adv-F/RGD and Adv-F/wt was observed in 293 cells, which show high-level expression of CAR. In this study, we attempted to apply Adv-F/RGD for gene therapy for malignant melanoma. At the same multiplicity of infection, melanoma cells infected with Adv-F/RGD carrying human interleukin 2 (AxCAhIL2-F/RGD) produced a higher level of cytokine than cells infected with AxCAhIL2-F/wt. Treatment by intratumoral injection of AxCAhIL2-F/RGD was more effective than intratumoral injection of AxCAhIL2-F/wt in regressing tumors in a melanoma xenograft model. These data suggest that integrin-targeted adenoviral vectors may be a powerful tool in gene therapy for CAR-deficient melanomas.
Collapse
Affiliation(s)
- Takafumi Nakamura
- Department of Molecular Medicine, Sapporo Medical University, S1 W17, Chuo-ku, Sapporo 060-8556, Japan
| | | | | |
Collapse
|