Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Gene Ther ; 21(3): 337-42, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24451117

ABSTRACT

Adoptive immunotherapy of tumors with T cells specific for the cancer-testis antigen NY-ESO-1 has shown great promise in preclinical models and in early stage clinical trials. Tumor persistence or recurrence after NY-ESO-1-specific therapy occurs, however, and the mechanisms of recurrence remain poorly defined. In a murine xenograft model of NY-ESO-1(+) multiple myeloma, we observed tumor recurrence after adoptive transfer of CD8(+) T cells genetically redirected to the prototypic NY-ESO-1157-165 peptide presented by HLA-A*02:01. Analysis of the myeloma cells that had escaped from T-cell control revealed intact expression of NY-ESO-1 and B2M, but selective, complete loss of HLA-A*02:01 expression from the cell surface. Loss of heterozygosity (LOH) in the major histocompatibility complex (MHC) involving the HLA-A locus was identified in the tumor cells, and further analysis revealed selective loss of the allele encoding HLA-A*02:01. Although LOH involving the MHC has not been described in myeloma patients with persistent or recurrent disease after immune therapies such as allogeneic hematopoietic cell transplantation (HCT), it has been described in patients with acute myelogenous leukemia who relapsed after allogeneic HCT. These results suggest that MHC loss should be evaluated in patients with myeloma and other cancers who relapse after adoptive NY-ESO-1-specific T-cell therapy.


Subject(s)
HLA-A2 Antigen/genetics , Heterozygote , Immunotherapy , Multiple Myeloma/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Tumor Escape , Alleles , Animals , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/therapy
3.
Tissue Antigens ; 81(4): 183-93, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23510414

ABSTRACT

The intrinsic anti-leukemic effect of allogeneic hematopoietic cell transplantation (HCT) is dependent on genetic disparity between donor and recipient, intimately associated with graft-versus-host disease (GVHD), and mediated by lymphocytes contained in or derived from the donor hematopoietic cell graft. Three decades of intense effort have not identified clinical strategies that can reliably separate the graft-versus-leukemia (GVL) effect from the alloimmune reaction that drives clinical GVHD. For patients who require HCT and for whom two or more human leukocyte antigen (HLA)-A, -B, -C, and -DRB1-matched donor candidates can be identified, consideration of donor and recipient genotype at additional genetic loci both within and outside the major histocompatibility complex may offer the possibility of selecting the donor [candidate(s)] that poses the lowest probability of GVHD and the highest probability of a potent GVL effect. Strategies for engineering conventional donor lymphocyte infusion also hold promise for prevention or improved treatment of post-transplant relapse. The brightest prospects for selectively enhancing the anti-leukemic efficacy of allogeneic HCT, however, are likely to be interventions that are designed to enhance specific antitumor immunity via vaccination or adoptive cell transfer, rather than those that attempt to exploit donor alloreactivity against the host. Adoptive transfer of donor-derived T cells genetically modified for tumor-specific reactivity, in particular, has the potential to transform the practice of allogeneic HCT by selectively enhancing antitumor immunity without causing GVHD.


Subject(s)
Genetic Loci/immunology , Graft vs Host Disease/prevention & control , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Leukemia/therapy , Chromosome Mapping , HLA Antigens/immunology , Histocompatibility , Humans , Leukemia/immunology , Leukemia/pathology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tissue Donors , Transplantation, Homologous
5.
Bone Marrow Transplant ; 40(10): 965-71, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17846603

ABSTRACT

Allogeneic hematopoietic cell transplantation (HCT) represents a potentially curative approach for patients with myelodysplastic syndromes (MDSs). While a large proportion of HCT recipients become long-term disease-free survivors, recurrence of MDS remains the leading cause of mortality after HCT. The role of donor lymphocyte infusion (DLI) in patients with relapsed MDS after HCT is unclear. We report results among 16 patients treated with DLI for relapsed MDS after HCT at a single institution between March 1993 and February 2004. The cohort contained 10 men and 6 women with a median age of 49 (range, 22-67) years. CR with resolution of cytopenias and prior disease markers occurred in 3 of 14 patients who could be evaluated. Two patients survived without MDS for 68 and 65 months after DLI, respectively, but died with pneumonia. Grades II-IV acute GVHD and chronic GVHD occurred after DLI in 6 (43%) and 5 (36%) patients, respectively. All three responders developed grades III-IV acute GVHD and extensive chronic GVHD after DLI. Our results confirm prior reports that DLI can result in CR in some patients with recurrent MDS after transplant, but long-term survival is infrequent.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Lymphocyte Transfusion , Myelodysplastic Syndromes/therapy , Adult , Aged , Female , Graft vs Host Disease/etiology , Humans , Lymphocyte Transfusion/adverse effects , Male , Middle Aged , Recurrence
6.
Leukemia ; 20(10): 1701-5, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16888618

ABSTRACT

Numerous reduced-intensity conditioning regimens for allogeneic hematopoietic cell transplantation are currently being explored, primarily in older patients and in individuals with comorbid conditions who are not eligible for conventional myeloablative conditioning regimens. There is agreement that these approaches have reduced early transplant-related (non-relapse) toxicity and mortality. It is unclear, however, whether these strategies improve long-term survival. Furthermore, as most trials with reduced-intensity regimens have enrolled older patients and patients with comorbid conditions, it is not appropriate to compare the results of these trials to those obtained with more conventional approaches. It remains to be determined whether younger patients, and patients without comorbid conditions, will derive significant long-term benefits from reduced-intensity regimens when compared to conventional strategies. It may be that the different approaches are complementary and in the end will preferentially serve specific patient populations based on age, comorbid conditions and malignancy type. To determine the role of reduced-intensity approaches, controlled prospective trials are needed, with enrolled patients being stratified according to comorbid conditions, disease characteristics, pre-transplant therapy and source of stem cells, at a minimum.


Subject(s)
Hematopoietic Stem Cell Transplantation , Medical Oncology/standards , Neoplasms/therapy , Transplantation Conditioning/methods , Transplantation Conditioning/standards , Humans , Neoplasms/mortality , Transplantation, Homologous
8.
Int J Hematol ; 76 Suppl 2: 155-61, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12430918

ABSTRACT

Immune-mediated elimination of tumor cells by donor T cells recognizing recipient minor H antigens contributes to the curative potential of allogeneic HCT. The importance of the allogeneic response to a successful outcome is clearly illustrated by the results of stem cell transplant for malignancy after nonmyeloablative conditioning. Remarkably little is understood about the molecular nature of minor H antigens and this has impeded efforts to determine the role of specific disparities in graft versus tumor reactions or to manipulate T cell responses to augment antitumor activity without exacerbating GVHD. The isolation of minor H antigen-specific CD8+ and CD4+ T cell clones from recipients of allogeneic HCT has provided the reagents to characterize their expression on leukemic progenitors and to identify the genes encoding these antigens. Using cDNA expression cloning, genetic polymorphisms in the human IFI-75, Uty, KIAA0020, and UGT2B17 genes have been identified to encode new minor H antigens presented by HLA A3, B8, A2, and A29 respectively. Two of these genes are preferentially expressed in hematopoietic cells including leukemic progenitors suggesting it may be possible to augment T cell responses to promote a selective graft versus leukemia effect. A third gene, UGT2B17 is highly expressed in liver and GI tract and may be a target for GVHD in these organs. The studies to identify the molecular nature of minor H antigens have provided insights into the complexities of the graft versus host response associated with allogeneic HCT, but the challenge for the future will be to develop strategies that can selectively induce durable graft versus tumor effects without GVHD. A critical issue in developing specific immunotherapy to augment GVL responses is to determine which minor H antigens are expressed on leukemic stem cells. Studies using transplantation of human AML into SCID mice have identified a putative leukemic stem cell which is contained in the CD34+ CD38- subset of the blast population and is present in very low frequency (<1/200,000) in blood or bone marrow from AML patents. We have examined the ability of minor H antigen-specific CTL to prevent engraftment of human AML in NOD/SCID mice. These studies show that engraftment of leukemias derived from individuals encoding the minor H antigen can be specifically prevented demonstrating that AML stem cells express minor H antigens and are targets for CTL. One approach to determine directly which minor H antigens can be selectively targeted to induce a GVL effect without GVHD is to adoptively transfer T cell clones of defined specificity and function to patients who relapse after HCT. Studies of this approach are now in progress in acute leukemia and have provided important insights into potential obstacles of T cell therapy for relapsed leukemia after HCT.


Subject(s)
Graft vs Leukemia Effect/immunology , Minor Histocompatibility Antigens/immunology , Animals , Antigens, Neoplasm/immunology , Epitopes, T-Lymphocyte , Humans
9.
Tissue Antigens ; 59(4): 293-303, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12135428

ABSTRACT

We have evaluated the utility of genetic linkage analysis to identify genes that encode minor histocompatibility antigens using vaccinia virus vectors as a simple and convenient method for transient expression of class I MHC molecules in lymphoblastoid cell lines. As a test case, we used a CTL clone that recognizes HA-8, a minor histocompatibility antigen encoded by the KIAA0020 gene and presented by HLA-A*0201. EBV-transformed B cell lines from individuals in three large pedigrees from the CEPH reference family collection were infected with a recombinant vaccinia virus vector encoding an HLA-A*0201 transgene, which led to high level expression of the MHC restricting allele HLA-A*0201 on the cell surface. HA-8 expression in the vaccinia-infected target cells was then determined using standard in vitro cytotoxicity assays. Pairwise linkage analysis of the segregation of HA-8 expression in these pedigrees demonstrated that the HA-8 gene was tightly linked with a cluster of marker loci located on the distal portion of chromosome 9p. Analysis of 9p marker haplotypes for individuals in the three families identified several individuals with recombinant haplotypes, and these recombination events were used to refine the precision of the HA-8 gene localization further. The data collectively indicate that the HA-8 gene is localized to a 10.3 cM (corresponding to 3.9 Mb) interval of distal 9p that is thought to encode at least 11 genes, including KIAA0020. These results demonstrate that linkage analysis can be used to map minor histocompatibility genes with high precision and accuracy. Over the next years, refinement and annotation of the human genome sequence will undoubtedly increase the utility of linkage analysis as a tool for identifying minor histocompatibility antigen genes.


Subject(s)
Chromosomes, Human, Pair 9 , HLA-A Antigens/genetics , Lod Score , Minor Histocompatibility Antigens/genetics , Antigens, Surface/genetics , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Transformed , Chromosome Mapping , Epitopes/genetics , Genetic Markers , HLA-A2 Antigen/genetics , Herpesvirus 4, Human/genetics , Humans , Phenotype , Polymorphism, Genetic
10.
Article in English | MEDLINE | ID: mdl-11722994

ABSTRACT

This article discusses changes in the way hematopoietic stem cell allotransplants may be carried out in the future to treat patients with malignant hematological diseases. Specifically, the focus has shifted away from attempts at eradicating underlying diseases through toxic high-dose chemoradiation therapy towards using the stem cell donor's immune cells for that purpose (allogeneic graft-versus-tumor effect). The non-myeloablative transplant approaches hold promise in reducing the morbidity and mortality associated with conventional high-dose chemoradiation therapy, and they allow allogeneic transplants in elderly or medically infirm patients who are at present not candidates for transplantation. In the future, specific graft-versus-tumor responses may become possible by eliciting donor T cell responses to tumor-associated minor histocompatibility antigens. In Section I, Dr. Rainer Storb describes experimental studies in random-bred dogs that rely on non-cytotoxic immunosuppressive agents to establish stable allografts. Powerful postgrafting immunosuppression, traditionally directed at preventing graft-versus-host disease (GVHD), is also used to overcome host-versus-graft (HVG) reactions, thereby dramatically reducing the need for intensive immunosuppressive conditioning programs. Preclinical canine studies have been translated into the clinical setting for treatment of elderly or medically infirm patients with malignant hematological diseases. The pretransplant conditioning has been reduced to a single dose of 2 Gy total body irradiation (TBI) with or without fludarabine. The lack of toxicity makes it possible for transplants to be conducted in the outpatient setting. Multicenter trials have been initiated, and more than 300 patients have been successfully treated with hematopoietic stem cell grafts both from related and unrelated HLA-matched donors. In Section II, Dr. Richard Champlin describes clinical studies with therapeutic strategies that utilize relatively non-toxic, nonmyeloablative disease-specific preparative regimens incorporating fludarabine, together with other chemotherapeutic agents, to achieve disease suppression and engraftment of allogeneic hematopoietic cells and to allow subsequent infusions of donor lymphocytes. Remissions have been seen in patients with acute myelocytic, chronic myelocytic, chronic lymphocytic, leukemias, lymphomas, and myelomas. In Section III, Dr. Stanley Riddell and colleagues describe studies on isolation of T cells reactive with minor histocompatibility (H) antigens and involved both in GVHD and graft-versus-leukemia (GVL) responses. For example, the gene encoding a novel H-Y antigen in humans has been identified and shown to exhibit restricted tissue expression. Acute myelocytic leukemia stem cells were demonstrated to express the H-Y antigen and additional minor H antigens, and engraftment of such cells in NOD/SCID mice could be selectively prevented by minor antigen-specific T-cell clones. An autosomal encoded human minor H antigen associated with chronic GVHD has been demonstrated. A trial evaluating therapy of relapsed acute myelocytic leukemia or acute lymphoblastic leukemia after allogeneic stem cell transplantation with T-cell clones specific for recipient minor H antigens has been initiated.


Subject(s)
Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Aged , Animals , Antigens, Neoplasm/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Humans , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/toxicity , Minor Histocompatibility Antigens/immunology , Transplantation, Homologous/immunology , Transplantation, Homologous/methods
12.
J Exp Med ; 193(2): 195-206, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11148223

ABSTRACT

Minor histocompatibility antigens (mHAgs) present a significant impediment to organ and bone marrow transplantation between HLA-identical donor and recipient pairs. Here we report the identification of a new HLA-A*0201-restricted mHAg, HA-8. Designation of this mHAg as HA-8 is based on the nomenclature of Goulmy (Goulmy, E. 1996. Curr. Opin. Immunol. 8:75-81). This peptide, RTLDKVLEV, is derived from KIAA0020, a gene of unknown function located on chromosome 9. Polymorphic alleles of KIAA0020 encode the alternative sequences PTLDKVLEV and PTLDKVLEL. Genotypic analysis demonstrated that the HA-8-specific cytotoxic T lymphocyte (CTL) clone SKH-13 recognized only cells that expressed the allele encoding R at P1. However, when PTLDKVLEV was pulsed onto cells, or when a minigene encoding this sequence was used to artificially translocate this peptide into the endoplasmic reticulum, it was recognized by CTLs nearly as well as RTLDKVLEV. This indicates that the failure of CTLs to recognize cells expressing the PTLDKVLEV-encoding allele of KIAA0020 is due to a failure of this peptide to be appropriately proteolyzed or transported. Consistent with the latter possibility, PTLDKVLEV and its longer precursors were transported poorly compared with RTLDKVLEV by transporter associated with antigen processing (TAP). These studies identify a new human mHAg and provide the first evidence that minor histocompatibility differences can result from the altered processing of potential antigens rather than differences in interaction with the relevant major histocompatibility complex molecule or T cell receptor.


Subject(s)
Antigen Presentation , Minor Histocompatibility Antigens/metabolism , Alleles , Amino Acid Sequence , Base Sequence , Clone Cells , DNA Primers/genetics , Epitopes/chemistry , Epitopes/genetics , Female , Humans , Male , Mass Spectrometry , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/genetics , Molecular Sequence Data , Pedigree , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
15.
J Immunol ; 164(5): 2807-14, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10679124

ABSTRACT

The mammalian Y chromosome encodes male-specific minor histocompatibility (H-Y) Ags that are recognized by female T cells in an MHC-restricted manner. Two human H-Y epitopes presented by HLA-A2 and HLA-B7, respectively, have been identified previously and both are derived from the SMCY gene. We previously isolated CD8+ CTL clones that recognized a male-specific minor histocompatibility Ag presented by HLA-B8. In contrast to the SMCY-encoded H-Y epitopes, the B8/H-Y Ag was not presented by fibroblasts from male donors, suggesting that it was encoded by a novel gene. We now report that the HLA-B8-restricted H-Y epitope is defined by the octameric peptide LPHNHTDL corresponding to aa residues 566-573 of the human UTY protein. Transcription of the UTY gene is detected in a wide range of human tissues, but presentation of the UTY-derived H-Y epitope to CTL by cultured human cells shows significant cell-type specificity. Identification of this CTL-defined H-Y epitope should facilitate analysis of its contribution to graft/host interactions following sex-mismatched organ and bone marrow transplantation.


Subject(s)
H-Y Antigen/genetics , HLA-B8 Antigen/genetics , Proteins/genetics , Amino Acid Sequence , Animals , COS Cells , Cell Line, Transformed , Cells, Cultured , Chromosome Mapping , Epitopes/biosynthesis , Epitopes/genetics , Female , Gene Expression Regulation/immunology , H-Y Antigen/biosynthesis , HLA-B8 Antigen/biosynthesis , Humans , Male , Minor Histocompatibility Antigens , Molecular Sequence Data , Nuclear Proteins , Oligopeptides/genetics , Oligopeptides/immunology , Oligopeptides/isolation & purification , Organ Specificity/genetics , Organ Specificity/immunology , Protein Biosynthesis , Proteins/immunology , Y Chromosome/immunology
16.
Qual Life Res ; 9(7): 873-82, 2000.
Article in English | MEDLINE | ID: mdl-11297030

ABSTRACT

The Patient Satisfaction with Asthma Medication (PSAM) questionnaire was developed because no treatment satisfaction questionnaire could be identified that was comprehensive yet brief enough for use in clinical trials. Adult moderate asthmatics residing in Canada using an inhaled medication (either salmeterol, formoterol, or albuterol) self-administered the questionnaire, which also included the Asthma Quality of Life Questionnaire (AQLQ). A total of 53 asthmatics (70% female, 45% married, mean age: 47 years) completed the questionnaire. Using variable clustering, four PSAM scales were identified: Inhaler Properties, Comparison with Other Medications, Overall Perception of Medication, and Relief. Internal-consistency reliability provided evidence of reliability and lack of redundancy (Cronbach's Alpha: 0.82-0.88). Test-retest reliability was acceptable (ICC values at or near 0.70). As expected, interscale PSAM correlations were moderate to high; correlations between the PSAM and the AQLQ were low to moderate. To assess known groups validity, respondents were categorized by self-reported degree of asthma control: 'very well controlled', 'somewhat controlled', and 'not well controlled'. Significant between-groups differences were found on all PSAM scales except Inhaler Properties. Patients categorized as 'very well controlled' tended to report highest PSAM scale scores. The PSAM questionnaire demonstrated reliability and validity in moderate asthmatics. Responsiveness should be assessed in future, prospective studies.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Patient Satisfaction , Surveys and Questionnaires , Adult , Female , Humans , Male , Middle Aged
17.
J Vasc Interv Radiol ; 10(10): 1293-303, 1999.
Article in English | MEDLINE | ID: mdl-10584643

ABSTRACT

OBJECTIVE: To determine the change in health-related quality of life associated with uterine fibroid embolization (UFE). MATERIALS AND METHODS: A health-related quality-of-life questionnaire was administered before and after therapy. The questionnaire contained validated scales from the Medical Outcomes Study, with additional domains and symptom items specific to fibroids. Patients treated with UFE for symptomatic uterine leiomyomata completed a health-related quality of life questionnaire before therapy. A follow-up quality of life questionnaire and an additional brief questionnaire to assess symptom improvement were completed 3 and 6 months postprocedure. Confirmatory reliability and validity testing was also conducted. Mean scores for each scale on the quality of life questionnaire were calculated and change scores were computed. RESULTS: Fifty women were enrolled in the study and completed the baseline assessment. Health-related quality of life scores improved in all instances at follow-up. Mean change scores were statistically significant for all domains between baseline and month 3 (P < .01) and between baseline and month 6 (P < .05) except backache (P = .12). CONCLUSION: Patients undergoing UFE report significant improvements in health-related quality of life and fibroid-specific symptoms. These findings suggest that the measurement of health-related quality of life may be an effective means of comparing the outcome of UFE with other fibroid therapies.


Subject(s)
Embolization, Therapeutic , Leiomyoma/therapy , Quality of Life , Uterine Neoplasms/therapy , Adult , Female , Humans , Leiomyoma/diagnosis , Middle Aged , Patient Satisfaction , Reproducibility of Results , Retrospective Studies , Surveys and Questionnaires , Uterine Neoplasms/diagnosis
18.
Proc Natl Acad Sci U S A ; 96(15): 8639-44, 1999 Jul 20.
Article in English | MEDLINE | ID: mdl-10411928

ABSTRACT

Effective immunotherapy for human leukemia based on infusions of T lymphocytes requires the identification of effector T cells that target the leukemic stem cell. The transplantation of human acute myeloid leukemia into nonobese diabetic/severe combined immune deficient (SCID) mice has identified a rare leukemic progenitor termed the SCID leukemia-initiating cell, which is present in low frequency in the leukemic population and is essential for establishing leukemic hematopoiesis. Thus, this transplant model may be ideally suited to identify effector T cells with antileukemic activity. We report that CD8(+) cytotoxic T lymphocyte (CTL) clones specific for minor histocompatibility antigens inhibit the engraftment of human acute myeloid leukemia cells in nonobese diabetic/SCID mice and demonstrate that this inhibition is mediated by direct CTL recognition of SCID leukemia-initiating cells. These results indicate that CD8(+) minor histocompatibility antigen-specific CTL may be mediators of the graft-versus-leukemia effect associated with allogeneic hematopoietic cell transplantation and provide an experimental model to identify and select T cell clones for immunotherapy to prevent or treat relapse after allogeneic hematopoietic cell transplantation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Leukemia, Myeloid/immunology , Minor Histocompatibility Antigens/immunology , Stem Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Transplantation , Clone Cells/immunology , Coculture Techniques , Cytotoxicity Tests, Immunologic , Flow Cytometry , Graft Rejection/immunology , Humans , Immunotherapy/methods , Mice , Mice, Inbred NOD , Mice, SCID
19.
Curr Opin Hematol ; 5(6): 429-33, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9814651

ABSTRACT

In allogeneic bone marrow transplantation, differences between donor and recipient at minor histocompatibility loci, which encode allelic proteins containing variant peptide sequences, may result in adverse immune reactions such as graft-versus-host disease (GVHD), or graft rejection, but also a beneficial graft versus leukemia (GVL) response. Some minor H antigens are restricted in their expression to hematopoietic cells, including leukemic progenitors, suggesting it may be possible to separate GVHD and GVL responses. The antigenic peptides for a few human minor H antigens have been identified, and efforts to identify the genes encoding minor H antigens are being pursued. These advances promise to provide a more detailed understanding of the immunobiology and pathogenesis of GVHD and GVL responses and opportunities to selectively augment T-cell responses that promote a GVL effect by adoptive immunotherapy with T-cell clones specific for defined minor H determinants.


Subject(s)
Bone Marrow Transplantation/immunology , Immunotherapy/methods , Major Histocompatibility Complex/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Clone Cells/immunology , Graft vs Tumor Effect , Humans , Leukemia/immunology , Leukemia/pathology , Major Histocompatibility Complex/genetics
SELECTION OF CITATIONS
SEARCH DETAIL