Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Intern Med ; 266(2): 141-53, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19623690

ABSTRACT

Interleukin-7 (IL-7) is required for the development and survival of T cells and plays a critical role in modulating T-cell homeostasis. This review will address current understanding of IL-7 biology, review recent clinical experiences and discuss potential future clinical applications of IL-7, or IL-7 blockade, in the setting of disease.


Subject(s)
Immunotherapy/methods , Interleukin-7/immunology , T-Lymphocytes/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , HIV Infections/immunology , HIV Infections/therapy , Homeostasis/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , Randomized Controlled Trials as Topic
2.
Bone Marrow Transplant ; 35 Suppl 1: S53-7, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15812532

ABSTRACT

Successful hematopoietic progenitor cell transplantation requires rapid and complete transfer of the donor hematopoietic and immune systems to the host. Whereas the uncontrolled transfer of a nontolerant donor immune system results in GVHD in many cases, strategies which diminish GVHD also diminish immune reconstitution. Thus, the reliable, rapid and safe transfer of immunity from donor to host remains a major challenge for the field. Advances in the understanding of the biology of immune reconstitution have elucidated that thymic-dependent immune reconstitution can restore global immunity, but is especially vulnerable to toxicities associated with transplant. Alternatively, homeostatic peripheral expansion can be exploited for targeted immunity toward pathogens and tumors, but is difficult to manipulate without exacerbating GVHD risk. New translatable strategies are needed to safely augment one or both of these pathways in the setting of allogeneic hematopoietic progenitor cell transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immune System , Recovery of Function , Animals , Graft Survival , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/trends , Humans , Transplantation, Autologous
5.
Hematol Oncol Clin North Am ; 15(4): 657-75, viii, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11676278

ABSTRACT

In the 3 decades since the signing of the National Cancer Act, there has been tremendous progress in the elucidation of the molecular underpinnings of cancer. Molecular genetic studies have been particularly insightful, revealing genetic rearrangements, such as chromosomal translocations, which may be the seminal event leading to deregulated cell growth for many childhood and adult cancers. These findings have led to new diagnostic and prognostic tools but have been slow to be translated into new therapeutic modalities. This article reviews a variety of methods now under development to exploit genetic changes in cancer to develop specific anticancer agents using gene therapy, viral therapy, and immunotherapy approaches. As many of these strategies inevitably enter the clinic, it will be imperative for health care professionals to be familiar with these novel approaches as they help patients navigate the likely broad array of treatment options.


Subject(s)
Neoplasms/therapy , Adenoviruses, Human/genetics , Adenoviruses, Human/physiology , Animals , Antigen Presentation , Child , Chromosome Aberrations , Clinical Trials as Topic , Defective Viruses/genetics , Defective Viruses/physiology , Drug Design , Forecasting , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , Genetic Therapy , Humans , Immune Tolerance , Immunotherapy , Mice , Models, Animal , Molecular Mimicry , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neoplasms/genetics , Nucleic Acid Conformation , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Oncogenes , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , RNA, Catalytic/therapeutic use , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , T-Lymphocyte Subsets/immunology , Transcription Factors/genetics , Transcription Factors/physiology
6.
J Neurosurg ; 95(2 Suppl): 270-5, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11599852

ABSTRACT

Molecular biological techniques have begun to transform modern medicine. These techniques have shown promise in the pathological diagnosis of difficult or uncommon tumors. Accurate molecular diagnosis of the small round-cell tumors, for example, is especially important because divergent therapies may be required to eradicate such disparate lesions as neuroblastoma, lymphoma, rhabdomyosarcoma, central primitive neuroectodermal tumors/medulloblastoma, or Ewing sarcoma (ES). The authors present an unusual case of a primary, extraosseous ES arising from the intramedullary spinal cord, in which molecular studies were required for specific diagnosis and therapeutic guidance.


Subject(s)
Cytogenetic Analysis , Sarcoma, Ewing/diagnosis , Spinal Cord Neoplasms/diagnosis , Adult , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/pathology , Sarcoma, Ewing/surgery , Spinal Cord Neoplasms/pathology , Spinal Cord Neoplasms/surgery
7.
Trends Immunol ; 22(10): 564-71, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11574281

ABSTRACT

Recent evidence has implicated interleukin-7 (IL-7) as a master regulator of T-cell homeostasis, based upon its essential role in the homeostatic expansion of naive T-cell populations in response to low-affinity antigens (Ags) and its capacity to enhance dramatically the expansion of peripheral T-cell populations in response to high-affinity Ags. Furthermore, T-cell-depleted humans have a unique inverse relationship between the peripheral CD4(+) T-cell count and the level of circulating IL-7. Together, these data suggest that increased amounts of IL-7 become available following T-cell depletion, thus, enhancing the high- and low-affinity Ag-driven expansion of the population of residual, mature T cells and boosting thymic regenerative capacity, as a means to restore T-cell homeostasis.


Subject(s)
Interleukin-7/immunology , T-Lymphocytes/immunology , Animals , Homeostasis/immunology , Humans , Lymphocyte Count , Lymphocyte Depletion , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/cytology
9.
J Clin Oncol ; 19(16): 3649-59, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11504746

ABSTRACT

PURPOSE: A propensity for hematogenous spread with resulting contamination of autologous cell products complicates cellular therapies for Ewing's sarcoma. We used a new approach to purge artificially contaminated cellular specimens of Ewing's sarcoma and show the capacity for real-time polymerase chain reaction (PCR) to quantify the contamination level of Ewing's sarcoma in such specimens. PATIENTS AND METHODS: Binding of monoclonal antibody (MoAb) 8H9 to Ewing's sarcoma cell lines and normal hematopoietic cells was studied using flow cytometry. Using real-time PCR--based amplification of t(11;22), levels of Ewing's contamination of experimental and clinical cellular products were monitored. Purging was accomplished using immunomagnetic-based depletion. Monitoring of the function of residual hematopoietic progenitors and T cells was performed using functional assays. RESULTS: MoAb 8H9 shows binding to Ewing's sarcoma but spares normal hematopoietic tissues. Nested real-time PCR is capable of detecting contaminating Ewing's sarcoma cells with a sensitivity of one cell in 10(6) normal cells. After 8H9-based purging, a 2- to 3-log reduction in contaminating Ewing's sarcoma was shown by real-time PCR, with purging to PCR negativity at levels of contamination of 1:10(6). Levels of contamination in clinical samples ranged from 1:10(5) to 10(6). Therefore, 8H9-based purging of clinical samples is predicted to reduce tumor cell contamination to a level below the limit of detection of PCR. CONCLUSION: These results demonstrate a new approach for purging contaminated cellular products of Ewing's sarcoma and demonstrate the capacity of real-time PCR to provide accurate quantitative estimates of circulating tumor burden in this disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunomagnetic Separation/standards , Sarcoma, Ewing/pathology , Sarcoma, Ewing/therapy , Animals , Bone Marrow/pathology , Bone Marrow Purging/standards , DNA Primers , Flow Cytometry , Humans , Leukapheresis/standards , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Sensitivity and Specificity , Tumor Cells, Cultured
11.
Cell Death Differ ; 8(5): 506-14, 2001 May.
Article in English | MEDLINE | ID: mdl-11423911

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is able to kill transformed cells. We have studied the expression and functionality of the TRAIL apoptotic pathway in Ewing's sarcoma. We demonstrate that tumors from patients with Ewing's sarcoma express receptors TRAIL-R1 and -R2. Using a panel of nine Ewing's sarcoma cell lines TRAIL could induce apoptosis in seven cell lines. Preincubation with interferon-gamma rendered the two resistant cell lines sensitive. TRAIL was the most potent inducer of apoptosis when compared to Fas ligand or TNF. TRAIL-mediated apoptosis could be inhibited by various caspase-inhibitors. No difference in the surface expression of TRAIL-receptors was observed between sensitive and resistant cell lines. Also, all cell lines had similar levels of expression of Flice-like inhibitory protein (FLIP) on immunoblot. However, the two resistant cell lines had only very low level expression of caspase 8 on RNA and protein level. In summary, we show that Ewing's sarcoma expresses receptors for TRAIL, and that cells are exquisitely sensitive to TRAIL-mediated apoptosis. These results may warrant clinical trials with TRAIL in Ewing's sarcoma once the safety of TRAIL for humans has been established.


Subject(s)
Apoptosis/drug effects , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/pharmacology , Sarcoma, Ewing/pathology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/genetics , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/genetics , Caspases/metabolism , Cycloheximide/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Drug Resistance , Flow Cytometry , Humans , Interferon-gamma/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , fas Receptor/metabolism
12.
Blood ; 97(10): 2983-90, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342421

ABSTRACT

Interleukin (IL)-7 is known to up-regulate thymopoietic pathways of T-cell regeneration. Recent work also has shown it to potently enhance thymic-independent peripheral expansion and to restore immunocompetence in athymic T-cell-depleted hosts. We hypothesized that endogenous IL-7 could contribute to the restoration of T-cell homeostasis following T-cell depletion. To analyze this, we evaluated circulating IL-7 levels and lymphocyte subsets in multiple clinical cohorts with T-cell depletion of varying etiologies. In pediatric (n = 41) and adult (n = 51) human immunodeficiency virus-infected CD4-depleted patients, there were strong inverse correlations between IL-7 levels and CD4 counts (r = -0.77, P <.0001, and r = -0.68, P <.0001). Declines in IL-7 were temporally correlated with recovery of CD4 counts. Similar patterns were observed in CD4-depleted patients receiving cancer chemotherapy (r = -0.65, P =.009). Therefore, in 2 disparate clinical scenarios involving CD4 depletion, IL-7 levels dynamically respond to changes in CD4 T-cell number, making this cytokine uniquely suited as a candidate regulator of T-cell homeostasis. Furthermore, in patients with idiopathic CD4 lymphopenia, a much weaker relationship between IL-7 levels and peripheral blood CD4 counts was observed, suggesting that an impaired IL-7 response to CD4 depletion may contribute to the impaired lymphocyte homeostasis observed in this population. In light of the known effects of IL-7 on T-cell regeneration, we postulate that increased availability of IL-7 could play a critical role in restoring T-cell homeostasis following T-cell depletion.


Subject(s)
HIV Infections/blood , Homeostasis , Interleukin-7/physiology , T-Lymphocytes/physiology , Adolescent , Adult , CD4 Lymphocyte Count , Child , Child, Preschool , Cohort Studies , HIV Infections/drug therapy , HIV Infections/immunology , HIV Protease Inhibitors/therapeutic use , Humans , Infant , Interleukin-7/blood , Longitudinal Studies , Lymphocyte Count , Lymphocyte Subsets , Ritonavir/therapeutic use
13.
J Clin Oncol ; 19(6): 1848-54, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11251017

ABSTRACT

There are major differences between therapeutic tumor vaccines and chemotherapeutic agents that have important implications for the design of early clinical trials. Many vaccines are inherently safe and do not require phase I dose finding trials. Patients with advanced cancers and compromised immune systems are not good candidates for assessing either the toxicity or efficacy of therapeutic cancer vaccines. The rapid pace of development of new vaccine candidates and the variety of possible adjuvants and modifications in method of administration makes it important to use efficient designs for clinical screening and evaluation of vaccine regimens. We review the potential advantages of a wide range of clinical trial designs for the development of tumor vaccines. We address the role of immunological endpoints in early clinical trials of tumor vaccines, investigate the design implications of attempting to use disease stabilization as an end point and discuss the difficulties of reliably utilizing historical control data. Several conclusions for expediting the clinical development of effective cancer vaccines are proposed.


Subject(s)
Antineoplastic Agents/therapeutic use , Cancer Vaccines/therapeutic use , Endpoint Determination , Neoplasms/virology , Randomized Controlled Trials as Topic , Adjuvants, Immunologic/therapeutic use , Cohort Studies , Humans , Immunocompromised Host , Neoplasms/drug therapy , Research Design , Treatment Outcome
14.
J Infect Dis ; 183(7): 1116-20, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11237839

ABSTRACT

Virologic and immunologic responses were examined for 33 human immunodeficiency virus (HIV)-infected children who participated for > or = 96 weeks in a phase 1/2 protocol of 16 weeks of indinavir monotherapy, followed by the addition of zidovudine and lamivudine. At week 96, a median increase of 199 CD4+ T cells/microL and a median decrease of 0.74 log(10) HIV RNA copies/mL were observed. The relationship between control of viral replication and CD4) T cell count was examined. Patients were categorized into 3 response groups on the basis of duration and extent of control of viral replication. Of 21 children with a transient decrease in virus load of > or = 0.7 log(10) HIV RNA copies/mL from baseline, 7 experienced sustained increases in CD4+, CD4+ CD45RA+, and CD4+ CD45RO+ T cell counts. CD4+ CD45RA+ (naive) T cells were the major contributor to CD4+ T cell expansion. Continued long-term immunologic benefit may be experienced by a subset of children, despite only transient virologic suppression.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , HIV , Indinavir/therapeutic use , Lamivudine/therapeutic use , Zidovudine/therapeutic use , Adolescent , CD4 Antigens/analysis , CD4 Lymphocyte Count , Child , Child, Preschool , Drug Therapy, Combination , Female , Follow-Up Studies , HIV/isolation & purification , HIV Infections/immunology , HIV Infections/virology , Humans , Leukocyte Common Antigens/analysis , Male , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , RNA, Viral/analysis , Viral Load
15.
Blood ; 97(6): 1525-33, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238086

ABSTRACT

Thymic-deficient hosts rely primarily on antigen-driven expansion to restore the peripheral T-cell compartment following T-cell depletion (TCD). The degree to which this thymic-independent pathway can restore immune competence remains poorly understood but has important implications for a number of clinical conditions including stem cell transplantation and human immunodeficiency virus (HIV) infection. A model of HY-mediated skin graft rejection by athymic, TCD mice was used to show that restoration of naive and recall responses via peripheral expansion requires transfer of only 25 x 10(6) lymph node (LN) cells representing approximately 10% of the T-cell repertoire. Constitutive expression of bcl-2 in the expanding inocula restored recall responses to HY at a substantially lower LN cell dose (1 x 10(6)), which is normally insufficient to induce HY-mediated graft rejection in athymic hosts. Interestingly, bcl-2 had no effect on primary responses. Interleukin-7 (IL-7) potently enhanced thymic-independent peripheral expansion and led to HY graft rejection using an LN cell dose of 1 x 10(6) in both primary and recall models. The restoration of immune competence by IL-7 appeared to be mediated through a combination of programmed cell death inhibition, improved costimulation, and modulation of antigen-presenting cell (APC) function. These results show that immune competence for even stringent antigens such as HY can be restored in the absence of thymic function and identify IL-7 as a potent modulator of thymic-independent T-cell regeneration.


Subject(s)
Immune System/drug effects , Immunocompromised Host/drug effects , Interleukin-7/immunology , T-Lymphocytes/cytology , Animals , Apoptosis/immunology , Cell Division/drug effects , Dendritic Cells/immunology , Female , Graft Rejection/immunology , Immune System/cytology , Immunization/adverse effects , Immunologic Memory , Interleukin-7/administration & dosage , Interleukin-7/pharmacology , Lymph Nodes/cytology , Lymph Nodes/transplantation , Lymphocyte Depletion/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Skin Transplantation/immunology , Thymectomy/adverse effects , Thymectomy/rehabilitation
16.
Blood ; 97(5): 1491-7, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222398

ABSTRACT

Thymic-dependent differentiation of bone marrow (BM)-derived progenitors and thymic-independent antigen-driven peripheral expansion of mature T cells represent the 2 primary pathways for T-cell regeneration. These pathways are interregulated such that peripheral T-cell expansion is increased in thymectomized versus thymus-bearing hosts after bone marrow transplantation (BMT). This study shows that this interregulation is due to competition between progeny of these 2 pathways because depletion of thymic progeny leads to increased peripheral expansion in thymus-bearing hosts. To test the hypothesis that competition for growth factors modulates the magnitude of antigen-driven peripheral expansion during immune reconstitution in vivo, a variety of T-cell active cytokines were administered after BMT. Of the cytokines (interleukins) tested (IL-3, IL-12, IL-6, IL-2, and IL-7), IL-2 modestly increased peripheral expansion in the face of increasing numbers of thymic emigrants, whereas IL-7 potently accomplished this. This report also demonstrates that the beneficial effect of IL-7 on immune reconstitution is related to both increases in thymopoiesis as well as a direct increase in the magnitude of antigen-driven peripheral expansion. Therefore, the administration of exogenous IL-7, and to a lesser extent IL-2, abrogates the down-regulation in antigen-driven peripheral expansion that occurs in thymus-bearing hosts after BMT. These results suggest that one mechanism by which T-cell-depleted hosts may support antigen-driven T-cell expansion in vivo is via an increased availability of T-cell-active cytokines to support clonal expansion.


Subject(s)
Bone Marrow Transplantation/methods , Interleukin-7/pharmacology , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Cell Differentiation/drug effects , Cytokines/pharmacology , Female , Hematopoiesis/drug effects , Interleukin-7/physiology , Leukocyte Common Antigens/analysis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Regeneration/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Thymus Gland/drug effects , Transplantation, Isogeneic , Up-Regulation/drug effects
17.
Cytotherapy ; 3(1): 19-29, 2001.
Article in English | MEDLINE | ID: mdl-12028840

ABSTRACT

BACKGROUND: There is growing interest in the use of dendritic cells (DCs) for treatment of malignancy and infectious disease. Our goal was to develop a clinical scale method to prepare autologous DCs for cancer clinical trials. METHODS: PBMC were collected from normal donors or cancer patients by automated leukapheresis, purified by counterflow centrifugal elutriation and placed into culture in polystyrene flasks at 1 x 10(6) cells/mL for 5-7 days at 37 degrees C, with 5% CO(2), with IL-4 and GM-CSF. Conditions investigated included media formulation, supplementation with heat in activated allogeneic AB serum or autologous plasma and time to harvest (Day 5 or Day 7). DCs were evaluated for morphology, quantitative yield, viability, phenotype and function, including mixed leukocyte response and recall response to tetanus toxoid and influenza virus. RESULTS: DCs with a typical immature phenotype (CD14-negative, CD1a-positive, mannose receptor-positive, CD80-positive, CD83-negative) were generated most consistently in RPMI 1640 supplemented with 10% allogeneic AB serum or 10% autologous plasma. Cell yield was higher at Day 5 than Day 7, without detectable differences in phenotype or function. In pediatric sarcoma patients, autologous DCs had enhanced function compared with monocytes from which they were generated. In this patient group, starting with 8.0 +/- 3.7 x 10(8) fresh or cryopreserved autologous monocytes, DC yield was 2.1 +/- 1.0 x 10(8) cells, or 29% of the starting monocyte number. DISCUSSION: In the optimized clinical-scale method, purified peripheral monocytes are cultured for 5 days in flasks at 1 x 10(6) cells/mL in RPMI 1640, 10% allogeneic AB serum or autologous plasma, IL-4 and GM-CSF. This method avoids the use of FBS and results in immature DCs suitable for clinical trials.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/transplantation , Immunotherapy/methods , Monocytes/cytology , Sarcoma/immunology , Sarcoma/therapy , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Size , Cell Survival/drug effects , Cell- and Tissue-Based Therapy/methods , Clinical Trials as Topic , Culture Media/chemistry , Culture Media/pharmacology , Dendritic Cells/drug effects , Flow Cytometry , Glucose/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Hydrogen-Ion Concentration , Interleukin-4/pharmacology , Lactic Acid/metabolism , Leukapheresis , Monocytes/drug effects , Time Factors , Transplantation, Autologous
18.
Curr Oncol Rep ; 2(6): 539-46, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11122890

ABSTRACT

Successful immune targeting of malignancies hinges upon the ability to activate specific T-cell populations to recognize and attack tumor but spare normal vital tissues. Investigators in the field of tumor immunology are currently utilizing at least three distinct approaches toward this goal. In the first approach, molecular targets of cytolytic T cells which spontaneously develop in tumor-bearing patients have been identified and are subsequently used as immunogens in immunotherapy trials. Whereas this approach originally focused upon the identification of tumor antigens in the immune-responsive tumors malignant melanoma and renal cell carcinoma, it surprisingly led to the identification of a variety of molecules that are now known to be expressed in other common pediatric and adult tumors. In the second approach, tumor-specific molecules (eg, mutant p53 and chromosomal translocations) that have been identified in individual tumors during the study of neoplastic transformation are used as immunogens. Because chromosomal translocations are common in pediatric tumors, such targets may be of particular interest in pediatric oncology. In the third approach, immunization with whole tumor cell components is undertaken with the assumption that the most immunogenic molecules within the tumor will dominate the immune response induced. The benefits and limitations for each approach, particularly as it pertains to the development of immunotherapy for pediatric tumors, are discussed in this article.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines , Immunotherapy/methods , Neoplasms/immunology , T-Lymphocytes/immunology , Antigens, Neoplasm/analysis , Child , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Humans , Neoplasms/therapy , Translocation, Genetic
19.
Blood ; 96(2): 754-62, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10887145

ABSTRACT

Administration of mobilized peripheral blood progenitor cells (PBPCs) after high-dose chemotherapy rapidly restores multilineage hematopoiesis, but the ability of such products to restore lymphocyte populations remains unclear. In this report, we evaluated immune reconstitution in a series of patients treated with sequential cycles of high-dose chemotherapy, followed by autologous PBPC infusions (median CD34(+) cell dose 7.2 x 10(6) cells/kg [range 2-29.3]). Although patients experienced rapid reconstitution of B cells and CD8(+) T cells, we observed CD4 depletion and diminished immune responsiveness in all patients for several months after completion of therapy. Mature CD4(+) T cells contained within the grafts did not appear to contribute substantially to immune reconstitution because CD4 counts did not differ between recipients of unmanipulated T-cell replete infusions versus CD34 selected, T-cell-depleted infusions. Rather, at 12 months after therapy, total CD4 count was inversely proportional to age (rho = -0.78, P =.04), but showed no relationship to CD34 cell dose (rho = -0.42, P =.26), suggesting that age-related changes within the host are largely responsible for the limited immune reconstitution observed. These results demonstrate that in the autologous setting, the infusion of large numbers of PBPCs is not sufficient to restore T-cell immune competence and emphasize that specific approaches to enhance immune reconstitution are necessary if immune-based therapy is to be used to eradicate minimal residual disease after autologous PBPC transplantation. (Blood. 2000;96:754-762)


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , CD4 Lymphocyte Count , Hematopoietic Stem Cell Transplantation , Adolescent , Adult , Aging , Antigens, CD34/analysis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes , CD8-Positive T-Lymphocytes , Child , Child, Preschool , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Immunity , Killer Cells, Natural , Lymphocyte Count , Neuroblastoma/drug therapy , Sarcoma/drug therapy , Transplantation, Autologous
20.
Stem Cells ; 18(1): 10-8, 2000.
Article in English | MEDLINE | ID: mdl-10661568

ABSTRACT

Although cancer itself is immunosuppressive, cytotoxic antineoplastic therapy is the primary contributor to the clinical immunodeficiency observed in cancer patients. The immunodeficiency induced by cytotoxic antineoplastic therapy is primarily related to T-cell depletion, with CD4 depletion generally more severe than CD8 depletion. Myeloablative therapy, dose-intensive alkylating agents, purine nucleoside analogs, and corticosteroids substantially increase the risk of therapy-induced immunosuppression. Restoration of T-cell populations following cytotoxic antineoplastic therapy is a complex process. Efficient recovery of CD4+ T cell populations requires thymic-dependent pathways which undergo an age-dependent decline resulting in prolonged CD4+ T-cell depletion in adults following T-cell-depleting therapy. Total CD8+ T-cell numbers recover in both children and adults relatively quickly post-therapy; however, CD8+ subset disruptions often remain for a prolonged period. The clinical management of patients with therapy-induced T-cell depletion involves the maintenance of a high index of suspicion for opportunistic pathogens, irradiation of blood products, prophylaxis for viral infections, and reimmunization in selected clinical circumstances. Future research avenues include efforts to rapidly rebuild immunity following cytotoxic antineoplastic therapy so that immune-based therapies may be utilized immediately following cytotoxic therapy to target minimal residual neoplastic disease.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxicity, Immunologic/immunology , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes/drug effects , Animals , Antineoplastic Agents/therapeutic use , Disease Management , Humans , Lymphocyte Count , Lymphocyte Depletion , Neoplasms/complications , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...