Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
J Clin Invest ; 133(10)2023 05 15.
Article En | MEDLINE | ID: mdl-36951958

BackgroundRefractory CMV viremia and disease are associated with significant morbidity and mortality in recipients of hematopoietic stem cell transplant (HCT).MethodsIn phase I/II trials, we treated 67 subjects for CMV viremia or disease arising after HCT with adoptive transfer of banked, third-party, CMVpp65-sensitized T cells (CMVpp65-VSTs). All were evaluable for toxicity and 59 for response. Evaluable subjects had CMV disease or persisting viremia that had failed at least 2 weeks of induction therapy with a median of 3 antiviral drugs; 84.7% had more than 3 of 11 high-risk features. CMVpp65-VSTs were specific for 1 to 3 CMVpp65 epitopes, presented by a limited set of HLA class I or II alleles, and were selected based on high-resolution HLA matching at 2 of 10 HLA alleles and matching for subject and subject's HCT donor for 1 or more alleles through which the CMVpp65-VSTs were restricted.ResultsT cell infusions were well tolerated. Of 59 subjects evaluable for response, 38 (64%) achieved complete or durable partial responses.ConclusionsRecipients responding to CMVpp65VSTs experienced an improved overall survival. Of the risk factors evaluated, transplant type, recipient CD4+ and CD8+ T cell levels prior to adoptive therapy, and the HLA restriction of CMVpp65-VSTs infused each significantly affected responses. In addition, CMVpp65-specific T cells of HCT donor or recipient origin contributed to the durability of both complete and partial responses.Trial RegistrationNCT00674648; NCT01646645; NCT02136797 (NIH).FundingNIH (P01 CA23766, R21 CA162002 and P30 CA008748); Aubrey Fund; Claire Tow Foundation; Major Family Foundation; "Rick" Eisemann Pediatric Research Fund; Banbury Foundation; Edith Robertson Foundation; Larry Smead Foundation.


Cytomegalovirus Infections , Hematopoietic Stem Cell Transplantation , Humans , Child , Cytomegalovirus , Viremia/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Cytomegalovirus Infections/therapy , Cytomegalovirus Infections/etiology , CD8-Positive T-Lymphocytes
2.
Blood Adv ; 6(16): 4859-4872, 2022 08 23.
Article En | MEDLINE | ID: mdl-35605246

We established and characterized a bank of 138 CMVpp65 peptide-specific T-cell (CMVpp65CTLs) lines from healthy marrow transplant donors who consented to their use for treatment of individuals other than their transplant recipient. CMVpp65CTL lines included 131 containing predominantly CD8+ T cells and 7 CD4+ T cells. CD8+ CMVpp65CTLs were specific for 1 to 3 epitopes each presented by one of only 34 of the 148 class I alleles in the bank. Similarly, the 7 predominantly CD4+ CMVpp65CTL lines were each specific for epitopes presented by 14 of 40 HLA DR alleles in the bank. Although the number of HLA alleles presenting CMV epitopes is low, their prevalence is high, permitting selection of CMVpp65CTLs restricted by an HLA allele shared by transplant recipient and hematopoietic cell transplant donor for >90% of an ethnogeographically diverse population of hematopoietic cell transplant recipients. Within individuals, responses to CMVpp65 peptides presented by different HLA alleles are hierarchical. Furthermore, within groups, epitopes presented by HLA B*07:02 and HLA A*02:01 consistently elicit immunodominant CMVpp65CTLs, irrespective of other HLA alleles inherited. All dominant CMVpp65CTLs exhibited HLA-restricted cytotoxicity against epitope loaded targets and usually cleared CMV infections. However, immunodominant CMVpp65CTLs responding to epitopes presented by certain HLA B*35 alleles were ineffective in lysing CMV-infected cells in vitro or controlling CMV infections post adoptive therapy. Analysis of the hierarchy of T-cell responses to CMVpp65, the HLA alleles presenting immunodominant CMVpp65 epitopes, and the responses they induce may lead to detailed algorithms for optimal choice of third-party CMVpp65CTLs for effective adoptive therapy.


Cytomegalovirus Infections , Hematopoietic Stem Cell Transplantation , Alleles , CD8-Positive T-Lymphocytes , Cytomegalovirus Infections/therapy , Epitopes , Humans , Immunodominant Epitopes
3.
Blood ; 135(21): 1870-1881, 2020 05 21.
Article En | MEDLINE | ID: mdl-32157281

Despite advances in T-cell immunotherapy against Epstein-Barr virus (EBV)-infected lymphomas that express the full EBV latency III program, a critical barrier has been that most EBV+ lymphomas express the latency I program, in which the single Epstein-Barr nuclear antigen (EBNA1) is produced. EBNA1 is poorly immunogenic, enabling tumors to evade immune responses. Using a high-throughput screen, we identified decitabine as a potent inducer of immunogenic EBV antigens, including LMP1, EBNA2, and EBNA3C. Induction occurs at low doses and persists after removal of decitabine. Decitabine treatment of latency I EBV+ Burkitt lymphoma (BL) sensitized cells to lysis by EBV-specific cytotoxic T cells (EBV-CTLs). In latency I BL xenografts, decitabine followed by EBV-CTLs results in T-cell homing to tumors and inhibition of tumor growth. Collectively, these results identify key epigenetic factors required for latency restriction and highlight a novel therapeutic approach to sensitize EBV+ lymphomas to immunotherapy.


Burkitt Lymphoma/therapy , Decitabine/pharmacology , Epigenesis, Genetic , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/isolation & purification , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Burkitt Lymphoma/genetics , Burkitt Lymphoma/immunology , Burkitt Lymphoma/virology , Cell Proliferation , Epstein-Barr Virus Infections/virology , Humans , Immunotherapy , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Viral Proteins/genetics , Viral Proteins/metabolism , Xenograft Model Antitumor Assays
4.
Oncoimmunology ; 8(7): 1570778, 2019.
Article En | MEDLINE | ID: mdl-31143508

Depletion of T regulatory cells (Tregs) in the tumor microenvironment is a promising cancer immunotherapy strategy. Current approaches for depleting Tregs are limited by lack of specificity and concurrent depletion of anti-tumor effector T cells. The transcription factor forkhead box p3 (Foxp3) plays a central role in the development and function of Tregs and is an ideal target in Tregs, but Foxp3 is an intracellular, undruggable protein to date. We have generated a T cell receptor mimic antibody, "Foxp3-#32," recognizing a Foxp3-derived epitope in the context of HLA-A*02:01. The mAb Foxp3-#32 selectively recognizes CD4 + CD25 + CD127low and Foxp3 + Tregs also expressing HLA-A*02:01 and depletes these cells via antibody-mediated cellular cytotoxicity. Foxp3-#32 mAb depleted Tregs in xenografts of PBMCs from a healthy donor and ascites fluid from a cancer patient. A TCRm mAb targeting intracellular Foxp3 epitope represents an approach to deplete Tregs.

5.
JCI Insight ; 3(4)2018 02 22.
Article En | MEDLINE | ID: mdl-29467338

EBV infection is associated with a number of malignancies of clinical unmet need, including Hodgkin lymphoma, nasopharyngeal carcinoma, gastric cancer, and posttransplant lymphoproliferative disease (PTLD), all of which express the EBV protein latent membrane protein 2A (LMP2A), an antigen that is difficult to target by conventional antibody approaches. To overcome this, we utilized phage display technology and a structure-guided selection strategy to generate human T cell receptor-like (TCR-like) monoclonal antibodies with exquisite specificity for the LMP2A-derived nonamer peptide, C426LGGLLTMV434 (CLG), as presented on HLA-A*02:01. Our lead construct, clone 38, closely mimics the native binding mode of a TCR, recognizing residues at position P3-P8 of the CLG peptide. To enhance antitumor potency, we constructed dimeric T cell engaging bispecific antibodies (DiBsAb) of clone 38 and an affinity-matured version clone 38-2. Both DiBsAb showed potent antitumor properties in vitro and in immunodeficient mice implanted with EBV transformed B lymphoblastoid cell lines and human T cell effectors. Clone 38 DiBsAb showed a stronger safety profile compared with its affinity-matured variant, with no activity against EBV- tumor cell lines and a panel of normal tissues, and was less cross-reactive against HLA-A*02:01 cells pulsed with a panel of CLG-like peptides predicted from a proteomic analysis. Clone 38 was also shown to recognize the CLG peptide on other HLA-A*02 suballeles, including HLA-A*02:02, HLA-A*02:04, and HLA-A*02:06, allowing for its potential use in additional populations. Clone 38 DiBsAb is a lead candidate to treat EBV malignancies with one of the strongest safety profiles documented for TCR-like mAbs.


Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Epstein-Barr Virus Infections/drug therapy , Neoplasms/drug therapy , Oncogene Proteins, Viral/antagonists & inhibitors , Viral Matrix Proteins/antagonists & inhibitors , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Blood Buffy Coat , CHO Cells , Cell Line, Tumor , Cricetulus , Cross Reactions , Crystallography, X-Ray , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , HEK293 Cells , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , HLA-A2 Antigen/metabolism , Herpesvirus 4, Human/immunology , Humans , Mice , Models, Molecular , Neoplasms/immunology , Neoplasms/virology , Oncogene Proteins, Viral/immunology , Peptide Library , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/immunology , Xenograft Model Antitumor Assays
6.
J Immunol ; 190(9): 4650-60, 2013 May 01.
Article En | MEDLINE | ID: mdl-23554313

NK cells are regulated by inhibiting and activating cell surface receptors. Most inhibitory receptors recognize MHC class I Ags and protect healthy cells from NK cell-mediated autoaggression. However, certain activating receptors, including the human activating killer cell Ig-like receptor (KIR) 2DS1, also recognize MHC class I. This fact raises the question of how NK cells expressing such activating receptors are tolerized to host tissues. We investigated whether the presence of HLA-C2, the cognate ligand for 2DS1, induces tolerance in 2DS1-expressing NK cells. Anti-HLA-C2 activity could be detected in vitro in some 2DS1 positive NK clones irrespective of the presence or absence of HLA-C2 ligand in the donor. The frequency of anti-HLA-C2 reactivity was high in donors homozygous for HLA-C1. Surprisingly, no significant difference was seen in the frequency of anti-HLA-C2 cytotoxicity in donors heterozygous for HLA-C2 and donors without HLA-C2 ligand. However, donors homozygous for HLA-C2, compared with all other donors, had significantly reduced frequency of anti-HLA-C2 reactive clones. The 2DS1 positive clones that express inhibitory KIR for self-HLA class I were commonly noncytotoxic, and anti-HLA-C2 cytotoxicity was nearly exclusively restricted to 2DS1 single positive clones lacking inhibitory KIR. 2DS1 single positive NK clones with anti-HLA-C2 reactivity were also present posttransplantation in HLA-C2 positive recipients of hematopoietic stem cell transplants from 2DS1 positive donors. These results demonstrate that many NK cells with anti-HLA-C2 reactivity are present in HLA-C1 homozygous and heterozygous donors with 2DS1. In contrast, 2DS1 positive clones from HLA-C2 homozygous donors are frequently tolerant to HLA-C2.


HLA-C Antigens/immunology , Immune Tolerance/immunology , Killer Cells, Natural/immunology , Receptors, KIR/immunology , Receptors, KIR/metabolism , Antigens, CD/immunology , Antigens, CD/metabolism , Down-Regulation/immunology , HLA-C Antigens/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Interleukin-15/immunology , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , Leukocyte Immunoglobulin-like Receptor B1 , Ligands , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Tissue Donors
7.
Blood ; 120(8): 1633-46, 2012 Aug 23.
Article En | MEDLINE | ID: mdl-22623625

The Wilms tumor protein (WT-1) is widely recognized as a tumor antigen that is expressed differentially by several malignancies. However, WT-1 peptides known to induce tumoricidal T cells are few. In the present study, we evaluated T-cell responses of 56 healthy donors to in vitro sensitization with autologous APCs loaded with a pool of overlapping 15-mer peptides spanning the sequence of WT-1. Thereafter, we mapped the WT-1 peptides eliciting responses in each individual, defined the immunogenic peptides, and identified their presenting HLA alleles. We report 41 previously unreported epitopes of WT-1: 5 presented by class II and 36 by class I alleles, including 10 that could be presented by more than 1 class I allele. IFNγ(+) T cells responding to 98% of the class I and 60% of the class II epitopes exhibited HLA-restricted cytotoxicity against peptide-loaded targets. T cells specific for 36 WT-1 peptides were evaluable for leukemocidal activity, of which 27 (75%) lysed WT-1(+) leukemic targets sharing their restricting HLA allele. Each epitope identified induced T-cell responses in most donors sharing the epitopes' presenting allele; these responses often exceeded responses to flanking peptides predicted to be more immunogenic. This series of immunogenic epitopes of WT-1 should prove useful for immunotherapies targeting WT-1(+) malignancies.


Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , Leukemia/immunology , Peptides/immunology , T-Lymphocytes/immunology , WT1 Proteins/immunology , Alleles , Amino Acid Sequence , Cell Line, Tumor , Epitope Mapping , HLA Antigens/genetics , Humans , Leukemia/genetics , Molecular Sequence Data , Peptides/chemistry , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , WT1 Proteins/chemistry
8.
Sci Transl Med ; 2(37): 37ra44, 2010 Jun 23.
Article En | MEDLINE | ID: mdl-20574068

The clinical symptomatology in the X-linked Wiskott-Aldrich syndrome (WAS), a combined immunodeficiency and autoimmune disease resulting from WAS protein (WASp) deficiency, reflects the underlying coexistence of an impaired T helper 1 (TH1) immunity alongside intact TH2 immunity. This suggests a role for WASp in patterning T(H) subtype immunity, yet the molecular basis for the TH1-TH2 imbalance in human WAS is unknown. We have discovered a nuclear role for WASp in the transcriptional regulation of the TH1 regulator gene TBX21 at the chromatin level. In primary TH1-differentiating cells, a fraction of WASp is found in the nucleus, where it is recruited to the proximal promoter locus of the TBX21 gene, but not to the core promoter of GATA3 (a TH2 regulator gene) or RORc (a TH17 regulator gene). Genome-wide mapping demonstrates association of WASp in vivo with the gene-regulatory network that orchestrates TH1 cell fate choice in the human TH cell genome. Functionally, nuclear WASp associates with H3K4 trimethyltransferase [RBBP5 (retinoblastoma-binding protein 5)] and H3K9/H3K36 tridemethylase [JMJD2A (Jumonji domain-containing protein 2A)] proteins, and their enzymatic activity in vitro and in vivo is required for achieving transcription-permissive chromatin dynamics at the TBX21 proximal promoter in primary differentiating TH1 cells. During TH1 differentiation, the loss of WASp accompanies decreased enrichment of RBBP5 and, in a subset of WAS patients, also of filamentous actin at the TBX21 proximal promoter locus. Accordingly, human WASp-deficient TH cells, from natural mutation or RNA interference-mediated depletion, demonstrate repressed TBX21 promoter dynamics when driven under TH1-differentiating conditions. These chromatin derangements accompany deficient T-BET messenger RNA and protein expression and impaired TH1 function, defects that are ameliorated by reintroducing WASp. Our findings reveal a previously unappreciated role of WASp in the epigenetic control of T-BET transcription and provide a new mechanism for the pathogenesis of WAS by linking aberrant histone methylation at the TBX21 promoter to dysregulated adaptive immunity.


Cell Nucleus/metabolism , Immunity/immunology , Th1 Cells/immunology , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/physiopathology , Actins/metabolism , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/genetics , Chromatin/metabolism , DNA/metabolism , Epigenesis, Genetic , Genetic Loci/genetics , Genome, Human/genetics , Histones/metabolism , Humans , Methylation , Mutant Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/cytology , Transcription, Genetic , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/deficiency
9.
Discov Med ; 8(43): 210-8, 2009 Dec.
Article En | MEDLINE | ID: mdl-20040272

The artificial antigen-presenting cells (AAPCs) described in this review were generated to facilitate the production of virus-specific T-cells for the treatment of infections in patients after bone marrow transplant. These AAPCs consist of murine 3T3 cells genetically modified to express critical human molecules needed for T-cell stimulation, such as the co-stimulatory molecules B7.1, ICAM-1, and LFA-3 and one of a series of 6 common HLA class I alleles. When T-cells were sensitized against cytomegalovirus (CMV) using AAPCs that express a shared HLA allele or using autologous antigen-presenting cells (APCs) loaded with the CMVpp65 antigen, they were activated and expanded to become HLA-restricted CMVpp65-specific T-cells. These T-cells demonstrated functional activity in vitro against CMV by producing IFN-gamma and inducing CMVpp65-specific cytotoxicity. T-cells sensitized with AAPCs recognized antigenic epitopes presented by each HLA allele known to be immunogenic in Man. Sensitization with AAPCs also permitted expansion of IFN-gamma+ cytotoxic T-cells against subdominant epitopes that were not effectively recognized by T-cells sensitized with autologous APCs. This panel of AAPCs provides a source of immediately accessible, standardizable, and replenishable "off the shelf" cellular reagents with the potential to make adoptive immunotherapy widely available for the treatment of lethal infections, cancer, and autoimmune diseases.


Antigen-Presenting Cells/metabolism , HLA Antigens/metabolism , Immunotherapy, Adoptive/methods , Antigen-Presenting Cells/immunology , Female , Humans , Male , Models, Biological
10.
J Immunol ; 183(4): 2837-50, 2009 Aug 15.
Article En | MEDLINE | ID: mdl-19635907

Adoptive transfer of virus-specific T cells can treat infections complicating allogeneic hematopoietic cell transplants. However, autologous APCs are often limited in supply. In this study, we describe a panel of artificial APCs (AAPCs) consisting of murine 3T3 cells transduced to express human B7.1, ICAM-1, and LFA-3 that each stably express one of a series of six common HLA class I alleles. In comparative analyses, T cells sensitized with AAPCs expressing a shared HLA allele or autologous APCs loaded with a pool of 15-mer spanning the sequence of CMVpp65 produced similar yields of HLA-restricted CMVpp65-specific T cells; significantly higher yields could be achieved by sensitization with AAPCs transduced to express the CMVpp65 protein. T cells generated were CD8(+), IFN-gamma(+), and exhibited HLA-restricted CMVpp65-specific cytotoxicity. T cells sensitized with either peptide-loaded or transduced AAPCs recognized epitopes presented by each HLA allele known to be immunogenic in humans. Sensitization with AAPCs also permitted expansion of IFN-gamma(+) cytotoxic effector cells against subdominant epitopes that were either absent or in low frequencies in T cells sensitized with autologous APCs. This replenishable panel of AAPCs can be used for immediate sensitization and expansion of virus-specific T cells of desired HLA restriction for adoptive immunotherapy. It may be of particular value for recipients of transplants from HLA-disparate donors.


Alleles , Cytomegalovirus/immunology , Histocompatibility Antigens Class I/genetics , Immunodominant Epitopes/immunology , Immunotherapy, Adoptive , Lymphocyte Activation/immunology , Phosphoproteins/immunology , T-Lymphocytes, Cytotoxic/transplantation , Viral Matrix Proteins/immunology , Animals , Cells, Cultured , Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/genetics , HLA-A2 Antigen , HLA-A24 Antigen , HLA-A3 Antigen , HLA-B Antigens/genetics , HLA-B7 Antigen , HLA-B8 Antigen , Histocompatibility Antigens Class I/biosynthesis , Humans , Immunodominant Epitopes/administration & dosage , Mice , NIH 3T3 Cells , Phosphoproteins/administration & dosage , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , T-Lymphocyte Subsets/virology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , Viral Matrix Proteins/administration & dosage
11.
J Immunol ; 179(2): 854-68, 2007 Jul 15.
Article En | MEDLINE | ID: mdl-17617576

The inhibitory 2DL1 and activating 2DS1 killer Ig-like receptors (KIR) both have shared ligand specificity for codon sequences in the C2 group HLA-Cw Ags. In this study, we have investigated NK cell activation by allogeneic target cells expressing different combinations of the HLA-KIR ligand groups C1, C2, and Bw4. We demonstrate that fresh NK cells as well as IL-2-propagated NK cells from 2DS1-positive donors that are homozygous for the C1 ligand group are activated in vitro by B lymphoblastoid cell lines expressing the C2 group. This response is, in part, due to the absence of C1 group recognition mediated by the inhibitory receptor 2DL2/3. This "missing self" alloresponse to C2, however, is rarely observed in NK cells from donors lacking 2DS1. Even in presence of 2DS1, the NK alloresponse is dramatically reduced in donors that have C2 group as "self." Analysis of selected NK clones that express 2DS1 mRNA and lack mRNA for 2DL1 demonstrates that activation by the C2 ligand and mAb cross-linking of 2DS1 in these clones induces IFN-gamma. Furthermore, this C2 group-induced activation is inhibited by Abs to both HLA class I and the receptor. Collectively, these studies demonstrate that NK cells from 2DS1-positive donors are activated by target cells that express the C2 group as an alloantigen. This leads to increased IFN-gamma-positive fresh NK cells and induces NK allocytotoxicity in IL2-propagated polyclonal NK cells and NK clones. This study also provides support for the concept that incompatibility for the HLA-KIR ligand groups C1, C2, and Bw4 dominates NK alloactivation in vitro.


Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/immunology , Animals , Cytotoxicity, Immunologic , HLA Antigens/immunology , Humans , Mice , Receptors, KIR , Receptors, KIR2DL1 , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Homologous
12.
Hum Immunol ; 68(1): 30-40, 2007 Jan.
Article En | MEDLINE | ID: mdl-17207710

The allelic diversity and associated human leukocyte antigen (HLA) disparity of 1775 bone marrow recipients and their unrelated donors, matched for six of six (1361/1775,77%), five of six (397/1775, 22%), or four of six (17/1775, 1%) HLA-A, -B, -DR antigens, were retrospectively evaluated. The comprehensive HLA analysis included the class I (A, B, C) and II (DRB1, DQA1, DQB1, DPA1, DPB1) loci. Most (>66%) of the predominantly Caucasian study population carried one or two of five to seven common alleles at each HLA locus. In spite of this limited diversity, 29% of the six of six antigen-matched transplants carried allele mismatches at HLA-A, -B, and/or -DRB1, and 92% carried at least one allele mismatch at one of the eight HLA loci tested. Of the 968 HLA-A,-B,-DRB1 allele-matched pairs, 89% carried mismatches at other HLA loci, predominantly at DP loci. The substantially greater than expected HLA allelic disparity between donor and recipient suggests extensive haplotypic diversity and underscores the importance of enhancing approaches to mitigate the deleterious effect of HLA mismatches.


Alleles , Bone Marrow Transplantation/immunology , Genetic Variation , HLA Antigens/genetics , Tissue Donors , HLA-D Antigens/genetics , Histocompatibility Antigens Class I/genetics , Histocompatibility Testing , Humans , Retrospective Studies
13.
J Immunol ; 172(5): 3289-96, 2004 Mar 01.
Article En | MEDLINE | ID: mdl-14978137

MAGE-3 is the most commonly expressed cancer testis Ag and thus represents a prime target for cancer vaccines, despite infrequent natural occurrence of MAGE-3-specific immune responses in vivo. We report in this study the successful induction of Ab, CD8(+), and CD4(+) T cells in nonsmall cell lung cancer patients vaccinated with MAGE-3 recombinant protein. Two cohorts were analyzed: one receiving MAGE-3 protein alone, and one receiving MAGE-3 protein with adjuvant AS02B. Of nine patients in the first cohort, three developed marginal Ab titers and another one had a CD8(+) T cell response to HLA-A2-restricted peptide MAGE-3 271-279. In contrast, of eight patients from the second cohort vaccinated with MAGE-3 protein and adjuvant, seven developed high-titered Abs to MAGE-3, and four had a strong concomitant CD4(+) T cell response to HLA-DP4-restricted peptide 243-258. One patient simultaneously developed CD8(+) T cells to HLA-A1-restricted peptide 168-176. The novel monitoring methodology used in this MAGE-3 study establishes that protein vaccination induces clear CD4(+) T cell responses that correlate with Ab production. This development provides the framework for further evaluating integrated immune responses in vaccine settings and for optimizing these responses for clinical benefit.


Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Lipid A/analogs & derivatives , Lung Neoplasms/immunology , Neoplasm Proteins/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Amino Acid Sequence , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/blood , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Epitopes, T-Lymphocyte/immunology , Humans , Lipid A/administration & dosage , Lipid A/immunology , Lymphocyte Activation/immunology , Melanoma/immunology , Molecular Sequence Data , Saponins/administration & dosage , Saponins/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Vaccines, DNA/administration & dosage
14.
Proc Natl Acad Sci U S A ; 100(15): 8862-7, 2003 Jul 22.
Article En | MEDLINE | ID: mdl-12853579

NY-ESO-1 is one of the most immunogenic proteins described in human cancers, based on its capacity to elicit simultaneous antibody and CD8+ T cell responses in vivo. Although HLA class II restricted epitopes from NY-ESO-1 have been identified, no broad survey has yet established the status of natural CD4+ T cell responses in cancer patients in relation to CD8+ and antibody responses. We used a recently developed general strategy for monitoring CD4+ responses that overcomes the need for prior knowledge of epitope or HLA restriction to analyze a series of 31 cancer patients and healthy donors for the presence of CD4+ T cells to NY-ESO-1, and related this response to NY-ESO-1 expression in tumor cells and serum antibodies to NY-ESO-1. None of the 18 patients that tested seronegative for NY-ESO-1 had detectable CD4+ T cell responses. On the contrary, 11 of 13 cancer patients with serum antibodies to NY-ESO-1 had polyclonal CD4+ T cell responses directed against various known and previously undescribed NY-ESO-1 epitopes. NY-ESO-1 peptide 80-109 was the most immunogenic, with 10 of 11 patients responding to this peptide. We show here that 12-mer determinants from NY-ESO-1 eliciting a CD4+ T cell response were peptide 87-98 with promiscuous HLA class II presentation, peptide 108-119 restricted by HLA-DP4, and peptides 121-132 and 145-156, both shorter epitopes from previously described HLA-DR4 peptides, also presented by HLA-DR7. This study represents the next step in compiling a comprehensive picture of the adaptive immune response to NY-ESO-1, and provides a general strategy for analyzing the CD4+ T cell response to other tumor antigens eliciting a humoral immune response.


Antibodies, Neoplasm/blood , Antigens, Neoplasm , CD4-Positive T-Lymphocytes/immunology , Membrane Proteins , Neoplasms/immunology , Proteins/immunology , Amino Acid Sequence , Antigen Presentation , Antigens, Neoplasm/genetics , Epitopes/genetics , Female , Histocompatibility Antigens Class II/metabolism , Humans , Immunization , In Vitro Techniques , Lymphocyte Activation , Melanoma/immunology , Molecular Sequence Data , Ovarian Neoplasms/immunology , Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology
15.
J Immunol ; 169(9): 5118-29, 2002 Nov 01.
Article En | MEDLINE | ID: mdl-12391228

Killer Ig-like receptor (KIR) genes constitute a multigene family whose genomic diversity is achieved through differences in gene content and allelic polymorphism. KIR haplotypes containing a single activating KIR gene (A-haplotypes), and KIR haplotypes with multiple activating receptor genes (B-haplotypes) have been described. We report the evaluation of KIR gene content in extended families, sibling pairs, and an unrelated Caucasian panel through identification of the presence or absence of 14 KIR genes and 2 pseudogenes. Haplotype definition included subtyping for the expressed and nonexpressed KIR2DL5 variants, for two alleles of pseudogene 3DP1, and for two alleles of 2DS4, including a novel 2DS4 allele, KIR1D. KIR1D appears functionally homologous to the rhesus monkey KIR1D and likely arose as a consequence of a 22 nucleotide deletion in the coding sequence of 2DS4, leading to disruption of Ig-domain 2D and a premature termination codon following the first amino acid in the putative transmembrane domain. Our investigations identified 11 haplotypes within 12 families. From 49 sibling pairs and 17 consanguineous DNA samples, an additional 12 haplotypes were predicted. Our studies support a model for KIR haplotype diversity based on six basic gene compositions. We suggest that the centromeric half of the KIR genomic region is comprised of three major combinations, while the telomeric half can assume a short form with either 2DS4 or KIR1D or a long form with multiple combinations of several stimulatory KIR genes. Additional rare haplotypes can be identified, and may have arisen by gene duplication, intergenic recombination, or deletions.


Haplotypes/genetics , Immunoglobulin Variable Region/genetics , Multigene Family/immunology , Receptors, Immunologic/analysis , Receptors, Immunologic/genetics , Alleles , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Transformed , Centromere/chemistry , Centromere/genetics , Consanguinity , Female , Gene Frequency/immunology , Genetic Variation/immunology , Genomics/methods , Histocompatibility Testing/methods , Humans , Linkage Disequilibrium/immunology , Macaca mulatta , Male , Molecular Sequence Data , Polymerase Chain Reaction/methods , Polymorphism, Single-Stranded Conformational , Pseudogenes/immunology , Receptors, KIR , Sequence Homology, Amino Acid , Siblings , Telomere/chemistry , Telomere/genetics
16.
Hum Immunol ; 63(4): 295-300, 2002 Apr.
Article En | MEDLINE | ID: mdl-12039411

Various molecular methods in conjunction with serologic assays are used for clinical human leukocyte antigen (HLA) typing. Although serologic reagents detect most HLA-A and -B allospecificities, serologic HLA-C typing is hampered by the lack of informative antisera for many of the known HLA-C gene products. HLA antigens not detected by serology, but detected by molecular methods, are often referred to as "blank" antigens. Their lack of reactivity with antibodies in serological assays often reflects the presence of null alleles. The present study has characterized an HLA-Cw*04 allele (Cw*0409N) detected by DNA typing but not by serology. In cultured B-lymphoid 13W09501 cells carrying this Cw*04 null allele, isoelectric focusing analysis could not detect any component with a pattern compatible with that of the product of the HLA-Cw*0401 allele, but detected components reacting with an anti-HLA-Cw4 and Cw6 monoclonal antibody. Sequencing analysis of the full length HLA-Cw4 cDNA amplified from the cell line 13W095-01 revealed a base deletion at codon 365 in exon 7, resulting in a reading frame shift that added 32 amino acids at the C-terminal of the HLA-Cw4 heavy chain. These results indicate that the HLA-Cw*0409N allele may produce a putative long HLA-Cw4 heavy chain that is not expressed on the cell surface.


HLA-C Antigens/genetics , Alleles , Amino Acid Sequence , Base Sequence , DNA, Complementary , Female , Humans , Male , Molecular Sequence Data
...