Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Ann Oncol ; 29(3): 724-730, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29272364

ABSTRACT

Background: We previously demonstrated that brentuximab vedotin (BV) used as second-line therapy in patients with Hodgkin lymphoma is a tolerable and effective bridge to autologous hematopoietic cell transplantation (AHCT). Here, we report the post-AHCT outcomes of patients treated with second-line standard/fixed-dose BV and an additional cohort of patients where positron-emission tomography adapted dose-escalation of second-line BV was utilized. Patients and methods: Patients on the dose-escalation cohort received 1.8 mg/kg of BV intravenously every 3 weeks for two cycles. Patients in complete remission (CR) after two cycles received two additional cycles of BV at 1.8 mg/kg, while patients with stable disease or partial response were escalated to 2.4 mg/kg for two cycles. All patients, regardless of treatment cohort, proceeded directly to AHCT or received additional pre-AHCT therapy at the discretion of the treating physician based on remission status after second-line BV. Results: Of the 20 patients enrolled to the BV dose-escalation cohort, 8 patients underwent BV dose-escalation. BV escalation was well-tolerated, but no patients who were escalated converted to CR. Of 56 evaluable patients treated across cohorts, the overall response rate (ORR) to second-line BV was 75% with 43% CR. Twenty-eight (50%) patients proceeded directly to AHCT without post-BV chemotherapy, and a total of 50 patients proceeded to AHCT. Thirteen patients received consolidative post-AHCT therapy with either radiation, BV, or a PD-1 inhibitor. After AHCT, the 2-year progression-free survival (PFS) and overall survival were 67% and 93%, respectively. The 2-year PFS among patients in CR at the time of AHCT (n = 37) was 71% compared with 54% in patients not in CR (p = 0.12). The 2-year PFS in patients who proceeded to AHCT directly after receiving BV alone was 77%. Conclusions: Second-line BV is an effective bridge to AHCT that produces responses of sufficient depth to provide durable remission in conjunction with AHCT (clinicaltrials.gov: NCT01393717).


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Combined Modality Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Hodgkin Disease/therapy , Immunoconjugates/administration & dosage , Adolescent , Adult , Brentuximab Vedotin , Combined Modality Therapy/mortality , Drug Resistance, Neoplasm , Female , Hematopoietic Stem Cell Transplantation/mortality , Hodgkin Disease/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/therapy , Progression-Free Survival , Salvage Therapy/methods , Salvage Therapy/mortality , Transplantation, Autologous , Young Adult
2.
Leukemia ; 28(12): 2376-87, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24811343

ABSTRACT

Impaired Fas-mediated apoptosis is associated with poor clinical outcomes and cancer chemoresistance. Soluble Fas receptor (sFas), produced by skipping of exon 6, inhibits apoptosis by sequestering Fas ligand. Serum sFas is associated with poor prognosis of non-Hodgkin's lymphomas. We found that the alternative splicing of Fas in lymphomas is tightly regulated by a long-noncoding RNA corresponding to an antisense transcript of Fas (FAS-AS1). Levels of FAS-AS1 correlate inversely with production of sFas, and FAS-AS1 binding to the RBM5 inhibits RBM5-mediated exon 6 skipping. EZH2, often mutated or overexpressed in lymphomas, hyper-methylates the FAS-AS1 promoter and represses the FAS-AS1 expression. EZH2-mediated repression of FAS-AS1 promoter can be released by DZNeP (3-Deazaneplanocin A) or overcome by ectopic expression of FAS-AS1, both of which increase levels of FAS-AS1 and correspondingly decrease expression of sFas. Treatment with Bruton's tyrosine kinase inhibitor or EZH2 knockdown decreases the levels of EZH2, RBM5 and sFas, thereby enhancing Fas-mediated apoptosis. This is the first report showing functional regulation of Fas repression by its antisense RNA. Our results reveal new therapeutic targets in lymphomas and provide a rationale for the use of EZH2 inhibitors or ibrutinib in combination with chemotherapeutic agents that recruit Fas for effective cell killing.


Subject(s)
Lymphoma, B-Cell/blood , Lymphoma, B-Cell/genetics , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , fas Receptor/blood , fas Receptor/genetics , Adenine/analogs & derivatives , Alternative Splicing , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein , Fas Ligand Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Histones/metabolism , Humans , Introns , Lymphoma, B-Cell/metabolism , Models, Biological , Piperidines , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Protein Binding , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Leukemia ; 28(2): 329-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23765229

ABSTRACT

Immunomodulatory drugs (IMiDs) are effective therapeutic agents with direct inhibitory effects on malignant B- and plasma-cells and immunomodulatory effects on the T-cell activation. This dual function of IMiDs makes them appealing candidates for combination with a cancer vaccine. We investigated the immune stimulatory effects of lenalidomide, administrated to mice in doses, which provided comparable pharmacokinetics to human patients, on the potency of a novel fusion DNA lymphoma vaccine. The combination was curative in the majority of mice with 8d pre-established syngeneic A20 lymphomas compared with vaccine or lenalidomide alone and induced immune memory. In vivo depletion experiments established the requirement for effector CD8(+) and CD4(+) T cells in protective immunity. Unexpectedly, lenalidomide alone was also associated with reduced numbers of systemic myeloid-derived suppressor cell (MDSC) and regulatory T cell (Treg) in tumor-bearing but not naïve mice, an effect that was independent of simple tumor burden reduction. These results confirm and extend results from other models describing the effect of lenalidomide on enhancing T-cell immunity, highlight the potency of this effect, and provide a rationale for clinical application. Independently, a novel mechanism of action reversing tumor-induced immune suppression by MDSC is suggested.


Subject(s)
Cancer Vaccines/immunology , Immunologic Factors/pharmacology , Lymphoma/immunology , Lymphoma/pathology , Thalidomide/analogs & derivatives , Animals , Antibodies/immunology , Antibody Specificity/immunology , Antineoplastic Agents/pharmacology , Disease Models, Animal , Female , Humans , Immunologic Memory , Lenalidomide , Lymphoma/mortality , Lymphoma/therapy , Mice , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thalidomide/pharmacology , Tumor Burden/drug effects , Tumor Burden/immunology
4.
Bone Marrow Transplant ; 48(2): 269-77, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22773122

ABSTRACT

To enhance the therapeutic index of allogeneic hematopoietic SCT (HSCT), we immunized 10 HLA-matched sibling donors before stem cell collection with recipient-derived clonal myeloma Ig, idiotype (Id), as a tumor antigen, conjugated with keyhole limpet hemocyanin (KLH). Vaccinations were safe in donors and recipients. Donor-derived KLH- and Id-specific humoral and central and effector memory T-cell responses were detectable by day 30 after HSCT and were boosted by post-transplant vaccinations at 3 months in most recipients. One patient died before booster vaccinations. Specifically, after completing treatment, 8/9 myeloma recipients had persistent Id-specific immune responses and 5/9 had improvement in disease status. Although regulatory T cells increased after vaccination, they did not impact immune responses. At a median potential follow-up period of 74 months, 6 patients are alive, the 10 patients have a median PFS of 28.5 months and median OS has not been reached. Our results provide proof of principle that neoantigen and tumor antigen-specific humoral and cellular immunity could be safely induced in HSCT donors and passively transferred to recipients. This general strategy may be used to reduce relapse of malignancies and augment protection against infections after allogeneic HSCT.


Subject(s)
Antigens, Neoplasm/immunology , Hematopoietic Stem Cell Transplantation/methods , Immunization/methods , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Tissue Donors , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Epitopes , Female , HLA Antigens/immunology , Hemocyanins/administration & dosage , Hemocyanins/immunology , Humans , Immunity, Cellular/immunology , Immunoglobulin Idiotypes/administration & dosage , Immunoglobulin Idiotypes/immunology , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/surgery , Transplantation Immunology , Transplantation, Homologous
5.
Leukemia ; 26(9): 2114-23, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22425892

ABSTRACT

Bone destruction is a hallmark of multiple myeloma and affects more than 80% of patients. However, current therapy is unable to completely cure and/or prevent bone lesions. Although it is accepted that myeloma cells mediate bone destruction by inhibition of osteoblasts and activation of osteoclasts, the underlying mechanism is still poorly understood. This study demonstrates that constitutive activation of p38 mitogen-activated protein kinase in myeloma cells is responsible for myeloma-induced osteolysis. Our results show that p38 is constitutively activated in most myeloma cell lines and primary myeloma cells from patients. Myeloma cells with high/detectable p38 activity, but not those with low/undetectable p38 activity, injected into severe combined immunodeficient (SCID) or SCID-hu mice caused bone destruction. Inhibition or knockdown of p38 in human myeloma reduced or prevented myeloma-induced osteolytic bone lesions without affecting tumor growth, survival, or homing to bone. Mechanistic studies showed that myeloma cell p38 activity inhibited osteoblastogenesis and bone formation and activated osteoclastogenesis and bone resorption in myeloma-bearing SCID mice. This study elucidates a novel molecular mechanism-activation of p38 signaling in myeloma cells-by which myeloma cells induce osteolytic bone lesions, and indicates that targeting myeloma cell p38 may be a viable approach to treating or preventing myeloma bone disease.


Subject(s)
Bone Diseases/etiology , Multiple Myeloma/complications , Multiple Myeloma/enzymology , Osteolysis/etiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Blotting, Western , Bone Diseases/enzymology , Bone Diseases/pathology , Case-Control Studies , Cell Communication , Cell Proliferation , Humans , Immunoenzyme Techniques , Mice , Mice, SCID , Multiple Myeloma/pathology , Osteolysis/enzymology , Osteolysis/pathology , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
6.
Ann Oncol ; 23(6): 1640-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22015451

ABSTRACT

BACKGROUND: Tumor lysis syndrome (TLS) is a life-threatening disorder characterized by hyperuricemia and metabolic derangements. The efficacy of rasburicase, administered daily for 5 days, has been well established. However, the optimal duration of therapy is unknown in adults. PATIENTS AND METHODS: We evaluated the efficacy of rasburicase (0.15 mg/kg) administered as single dose followed by as needed dosing (maximum five doses) versus daily dosing for 5 days in adult patients at risk for TLS. RESULTS: Eighty of the 82 patients enrolled received rasburicase; 40 high risk [median uric acid (UA) 8.5 mg/dl; range, 1.5-19.7] and 40 potential risk (UA = 5.6 mg/dl; range, 2.4-7.4). Seventy-nine patients (99%) experienced normalization in their UA within 4 h after the first dose; 84% to an undetectable level (<0.7 mg/dl). Thirty-nine of 40 (98%) patients in the daily-dose arm and 34 of 40 (85%) patients in single-dose arm showed sustained UA response. Six high-risk patients within the single-dose arm required second dose for UA >7.5 mg/dl. Rasburicase was well tolerated; one patient with glucose-6-phosphate dehydrogenase deficiency developed methemoglobinemia and hemolysis. CONCLUSIONS: Rasburicase is highly effective for prevention and management of hyperuricemia in adults at risk for TLS. Single-dose rasburicase was effective in most patients; only a subset of high-risk patients required a second dose.


Subject(s)
Gout Suppressants/administration & dosage , Tumor Lysis Syndrome/prevention & control , Urate Oxidase/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Female , Gout Suppressants/therapeutic use , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Male , Middle Aged , Risk Factors , Treatment Outcome , Tumor Lysis Syndrome/etiology , Urate Oxidase/therapeutic use , Uric Acid/blood
7.
Vaccine ; 29(18): 3476-82, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21382485

ABSTRACT

Our laboratory previously described the strategy of fusing chemokine receptor ligands to antigens in order to generate immunogenic DNA vaccines. In the present study, we produced mouse ß-2 defensin (mBD2) fusion proteins using both ovalbumin (OVA) and gp100 as model antigens. Superior cross-presentation by dendritic cells (DC) was observed for mBD2 fused antigens over unfused antigens in vitro. In vivo, we observed significant increases in the expansion of adoptively transferred antigen-specific MHC class I, but not class II-restricted T cells after immunization with mBD2 fused antigen over antigen alone. This enhanced expansion of class I restricted T cells was Toll-like receptor 4 (TLR4) dependent, but CC chemokine receptor 6 (CCR6) independent. Superior tumor resistance was observed for mBD2-fusion protein vaccines, compared to unfused antigen, in both B16-OVA and B16 tumor models. These data suggest that production of mBD2 fusion proteins is feasible and that the vaccines facilitate in vivo expansion of adoptively transferred T cells through a TLR4-dependent mechanism.


Subject(s)
Adoptive Transfer , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Toll-Like Receptor 4/immunology , beta-Defensins/immunology , Animals , Antigen-Presenting Cells , Cross-Priming , Interferon-gamma/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Receptors, CCR6/immunology , Recombinant Fusion Proteins/immunology , gp100 Melanoma Antigen/immunology
8.
Ann Oncol ; 20(4): 709-14, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19150940

ABSTRACT

BACKGROUND: To determine the efficacy and side-effects of (90)Y ibritumomab tiuxetan (Zevalin) as front-line treatment in patients with early-stage extranodal indolent lymphoma of the ocular adnexa (orbit, conjunctiva, or eyelid). PATIENTS AND METHODS: From August 2004 to November 2007, 12 patients with stages I-E extranodal indolent lymphoma of the ocular adnexa were enrolled in a prospective trial of rituximab followed by (90)Y ibritumomab tiuxetan (Zevalin therapeutic regimen). For each patient, clinical examinations and imaging studies were used to document response to therapy using the The International Working Group response criteria. All patients had (111)In ibritumomab tixuetan imaging to confirm expected biodistribution before (90)Y-Zevalin therapy; in addition, three patients had an optional single photon emission computed tomography-computed tomography scan to estimate the absorbed radiation dose to the orbital and ocular tissues. RESULTS: The study included seven women and five men. The median age was 60 years (range 22-79). Nine patients had mucosa-associated lymphoid tissue lymphoma of conjunctiva or orbit; three patients had grades 1-2 follicular lymphoma of orbit. One patient who had been deemed stage I-E initially was found to have another lesion in her deltoid muscle on positron emission tomography 2 weeks after enrollment. She was kept on trial although her disease was reclassified as stage IV due to this single additional (biopsy-proven) site. Ten patients had a complete response and two partial response (PR) within 3 months of treatment. One patient had a recurrence in the upper eyelid 6 months after an initial PR; he then received 30 Gy of external-beam radiotherapy (EBRT). His disease later progressed again in the orbit and he is currently being considered for other treatments. A second patient who attained a PR has remained stable with no progression 12 months after treatment. With a median follow-up time of 20 months (range 6-44 months), there were no cases of distant (extraorbital) relapse. All 12 patients experienced grade I or II transient pancytopenia during the first 3 months after enrollment in the trial. There were no episodes of grade III or IV myelosuppression. The estimated absorbed radiation dose to the orbital soft tissues was <3 Gy, 10 times lower than that with EBRT. CONCLUSIONS: Rituximab followed by (90)Y ibritumomab tiuxetan is an effective and safe front-line treatment for early-stage extranodal indolent B-cell lymphoma of the ocular adnexa.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Eye Neoplasms/drug therapy , Lymphoma, B-Cell, Marginal Zone/drug therapy , Radioimmunotherapy , Yttrium Radioisotopes/therapeutic use , Adult , Aged , Antibodies, Monoclonal/adverse effects , Eye Neoplasms/pathology , Female , Humans , Lymphoma, B-Cell, Marginal Zone/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Prospective Studies , Yttrium Radioisotopes/adverse effects
9.
Leukemia ; 22(1): 179-85, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17898787

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive B-cell lymphoma with poor clinical outcome. Although front therapy induces a high rate of complete remission (CR), relapse is inevitable and new regimens are much needed for relapsed MCL. The proteasome inhibitor bortezomib (BTZ) induces apoptosis and sensitizes MCL cells to chemotherapy in relapsed MCL, but CR rates are low, with a short duration of response and severe toxicity. Here we evaluated whether BTZ is additive or synergistic with cyclophosphamide (CTX) and rituximab (RTX). Increasing doses of BTZ with a fixed dose of RTX and CTX (BRC regimen) resulted in markedly synergistic growth inhibition of MCL cells. BRC significantly enhanced apoptosis in MCL cell lines and primary tumor cells compared with single-agent treatment. Furthermore, western blotting analysis indicated that BRC induces apoptosis earlier via activation and cleavage of caspases-8, -9 and -3, and poly (ADP-ribose) polymerase, than single-agent treatment. The pan-caspase inhibitor completely blocked apoptosis induced by BRC. In vivo studies showed that BRC eradicated subcutaneous tumors in MCL-bearing SCID mice and significantly prolonged the long-term event-free survival in 70% of the mice. Hence, our study demonstrates that cytoreductive chemotherapy with both BTZ and anti-CD20 antibody may offer a better therapeutic modality for relapsed MCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Lymphoma, Mantle-Cell/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Blotting, Western , Boronic Acids/administration & dosage , Bortezomib , Caspases/metabolism , Cell Proliferation , Cyclophosphamide/administration & dosage , Drug Synergism , Immunophenotyping , In Vitro Techniques , Lymphoma, Mantle-Cell/drug therapy , Male , Mice , Mice, SCID , Poly(ADP-ribose) Polymerases/metabolism , Pyrazines/administration & dosage , Rituximab , Survival Rate , Transplantation, Heterologous
10.
Bone Marrow Transplant ; 36(4): 315-23, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15968284

ABSTRACT

The unique antigenic determinants (idiotype (Id)) of the immunoglobulin secreted by myeloma tumor can serve as a tumor-specific antigen for active immunotherapy. Our objective was to induce tumor-specific T-cell immunity in bone marrow transplant (BMT) donors to enhance antitumor effects of allografts. We vaccinated five HLA-matched sibling donors with myeloma Id proteins isolated from recipient plasma before bone marrow harvest. Recipients were administered booster Id immunizations following transplantation. Vaccination induced donor Id and carrier-specific cellular and/or humoral immune responses. Two recipients died within 30 days of BMT from transplant-related complications. Id and carrier-specific T-cell responses were detected in all three remaining patients post-, but not pre-BMT and persisted for 18 months. All three surviving patients converted from partial to complete responses following BMT. Two of the three patients remain disease-free 7 years and 8 years after BMT, and the third died of renal failure after 5.5 years while in complete remission from myeloma. Our results suggest that myeloma Id vaccination induces specific T-cell immunity in healthy donors which may be transferable by BMT, is associated with prolonged disease-free survival of recipients, and may represent a general strategy to enhance graft-versus-tumor effect in other malignancies for which defined tumor-specific antigens exist.


Subject(s)
Antigens, Neoplasm/administration & dosage , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation/methods , Immunization , Multiple Myeloma/therapy , Tissue Donors , Adult , Antigens, Neoplasm/pharmacology , Cancer Vaccines/therapeutic use , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation/mortality , Humans , Immunity/drug effects , Immunoglobulin Idiotypes/therapeutic use , Male , Middle Aged , Multiple Myeloma/mortality , Siblings , Survival Rate , T-Lymphocytes/immunology , Transplantation, Homologous , Treatment Outcome
11.
Ann Rheum Dis ; 62 Suppl 2: ii17-21, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14532141

ABSTRACT

A number of antimicrobial peptides such as defensins have multiple functions in host defence. Defensins are produced not only by phagocytic cells and lymphocytes, but also by the epithelial cell lining of the gastrointestinal and genitourinary tracts, the tracheobronchial tree, and keratinocytes. Some are produced constitutively, whereas others are induced by proinflammatory cytokines and exogenous microbial products. Defensins produced by cells in the course of innate host defence serve as signals which initiate, mobilise, and amplify adaptive immune host defences. Administration of defensins with antigens to mice enhances both cellular (Th1-dependent) and humoral (Th2-dependent) cytokine production and immune responses. Linkage of defensins to weak tumour antigens potentiates their immunoadjuvant effects. Defensins use multiple cellular receptors, which endows them with the capacity to marshall adaptive host defences against microbial invaders.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Defensins/immunology , Animals , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Cathelicidins , Chemotaxis/immunology , Dendritic Cells/immunology , Humans , Immunity, Cellular/immunology , Mice
12.
Biomed Pharmacother ; 56(3): 129-32, 2002 May.
Article in English | MEDLINE | ID: mdl-12046683

ABSTRACT

Multiple myeloma (MM) responds to, but is not cured by, chemotherapy and may therefore be amenable to tumor-specific immunization in the setting of minimal residual disease. The idiotype of the monoclonal immunoglobulin expressed by the tumor provides a clear tumor-specific antigen. Patients with follicular lymphoma have unequivocally established that idiotypic vaccination, administered when patients have minimal residual disease, has an antitumor effect and potential to improve the clinical outcome. This result and preclinical studies demonstrating that MM cells display idiotypic peptides on their surface in a form suitable for recognition and killing by host T cells, foster the application of idiotypic vaccination in MM. The current vaccine production involves idiotype protein purification for each patient followed by conjugation to exogenous, immunogenic carriers in order to break immunological tolerance. Furthermore, recent advances in molecular cloning and development of novel antigen delivery systems are making it possible to streamline the production of equally or more effective idiotypic vaccines. Particularly, DNA vaccines utilising genetic carriers to target idiotype on dendritic cells in vivo have proven successful in preclinical models. Additional candidate T cell antigens, such as MUC1, the cancer-testis antigens, and telomerase have been identified as potential targets for immunization. The possibility of using whole myeloma cells as a source of tumor antigens for immunotherapy is also being actively explored. Finally, clinical studies have begun in which dendritic cells are generated ex vivo, loaded with tumor antigen(s), and reinfused to immunize patients.


Subject(s)
Cancer Vaccines/immunology , Immunoglobulin Idiotypes/immunology , Immunotherapy , Multiple Myeloma/therapy , Cancer Vaccines/therapeutic use , Humans , Immunoglobulin Idiotypes/therapeutic use , Multiple Myeloma/immunology , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use
13.
J Immunol ; 167(11): 6644-53, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11714836

ABSTRACT

Chemokine receptors are differentially expressed on immature and mature dendritic cells (DC). Herein, we demonstrate for the first time that murine antimicrobial peptides beta-defensins 2 and 3 bind murine CCR6, similarly to inflammatory chemokine macrophage-inflammatory protein 3alpha, and they chemoattract bone marrow-derived immature, but not mature DC. Using various chemokines or defensins fused with nonimmunogenic tumor Ags, we studied their capacity to delivery Ags to subsets of immune cells to elicit antitumor immunity. We demonstrate that DNA immunizations with fusion constructs with beta-defensin 2 or inflammatory chemokines that target immature DC, but not homeostatic chemokines secondary lymphoid tissue chemokine, CCL21, or stromal cell-derived factor 1, CXCL12, which chemoattract mature DC, elicit humoral, protective, and therapeutic immunity against two different syngeneic lymphomas.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Vaccines, DNA/immunology , Animals , Antibodies, Anti-Idiotypic/biosynthesis , Antibodies, Anti-Idiotypic/genetics , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Chemokines/administration & dosage , Chemokines/genetics , Chemokines/immunology , Chemokines/physiology , Female , Gene Targeting , Humans , Immunity, Innate/genetics , Immunoglobulin Variable Region/administration & dosage , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Lymphoma, B-Cell/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Receptors, Chemokine/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/physiology , Tumor Cells, Cultured , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , beta-Defensins/administration & dosage , beta-Defensins/genetics , beta-Defensins/immunology , beta-Defensins/physiology
15.
Semin Hematol ; 38(3): 260-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11486314

ABSTRACT

The failure of chemotherapy to cure a significant proportion of multiple myeloma (MM) patients and increasing knowledge of tumor immunology and MM biology have generated considerable interest in immunotherapy for this lethal disease. Immunotherapy for MM can be divided into three broad categories: passive antibody-mediated immunotherapy, active specific immunization (vaccination), and adoptive T-cell immunotherapy. Early clinical trials using anti-CD20 monoclonal antibodies (mAbs) have met with limited success so far but have also suggested that selected patient subgroups may benefit from this treatment. The availability of a truly tumor-specific antigen such as immunoglobulin idiotype, the recent demonstration that MM cells process and present idiotype to T lymphocytes, and formal evidence of an antitumor effect of idiotypic vaccination in follicular lymphoma provide the framework for applying idiotypic vaccination in MM. The ability to generate ex vivo functional dendritic cells has made it possible to fuse them with patients' MM cells, thus producing a different type of customized vaccine. Dendritic cells are also a pivotal reagent to generate ex vivo MM-specific cytotoxic T lymphocytes (CTLs) to be reinfused into the patient for adoptive immunotherapy. This review summarizes achievements in MM immunotherapy based on data reported since 1998.


Subject(s)
Multiple Myeloma/therapy , Antigens, Neoplasm , Clinical Trials as Topic , Humans , Immunotherapy , Multiple Myeloma/immunology , Treatment Outcome
16.
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
17.
J Cancer Res Clin Oncol ; 127 Suppl 2: R14-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11768619

ABSTRACT

Keyhole limpet hemocyanin (KLH) has been used as an immune potentiator to enhance antigen-specific responses against haptens and weak antigens including self-antigens. In the present study, we describe the optimization of the intracellular cytokine response to KLH in peripheral blood mononuclear cells (PBMC) of lymphoma and myeloma patients that were vaccinated with tumor-specific immunoglobulin (Id) conjugated to KLH. Addition of anti-CD28 antibody significantly enhanced cytokine-producing CD4-T cells. While fresh PBMC showed maximal cytokine response 14 h after antigen stimulation, the frozen PBMC showed maximal cytokine responses by 24 h. Supplementation of the culture medium with fetal bovine serum gave a better signal-to-noise ratio than human AB serum in the intracellular detection of cytokines. The intracellular cytokine responses correlated with the cytokine measurements by enzyme-linked immunosorbent assay (ELISA). Together these results indicate that the intracellular cytokine assay is very helpful to measure antigen-specific immune responses, and in subsequent studies, we have utilized this sensitive technique to detect immune responses against tumor antigens such as idiotype.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cytokines/biosynthesis , Hemocyanins/immunology , Immunoglobulin Idiotypes/immunology , CD28 Antigens/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Vaccination , Vaccines, Conjugate/immunology
18.
Blood ; 96(8): 2828-33, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11023518

ABSTRACT

Immunoglobulin secreted by myeloma cells contains a unique antigenic determinant (idiotype [Id]) that may serve as a tumor-specific antigen. Although Id-protein-specific T-cell responses have been reported in patients with myeloma, it is not known whether primary myeloma tumor cells can present naturally processed Id peptides on their surface as a target. We immunized 2 healthy human stem-cell donors with Id proteins from their recipients. T cells from the immunized donors released high levels of T-helper 1-type cytokines in response to stimulation with myeloma cells from their recipients. The T-cell-mediated cytokine response to tumor cells was blocked by a major histocompatibility complex (MHC) class I monoclonal antibody, whereas the response to soluble Id protein was dependent on MHC class II. To investigate whether Id-specific CD8(+) T cells can recognize and kill autologous myeloma cells, we generated T cells from peripheral blood mononuclear cells from a third patient with myeloma by means of in vitro stimulation with autologous dendritic cells pulsed with Id protein. Tumor-specific lysis of myeloma cells was demonstrated by the lack of killing of autologous nonmalignant B cells or natural killer-sensitive K562 cells. Lysis of autologous myeloma targets was restricted by MHC class I molecules. These data represent the first report of class I-restricted T-cell recognition of fresh autologous myeloma targets and formally demonstrate that human myeloma cells can serve as targets of an Id-specific T-cell response. (Blood. 2000;96:2828-2833)


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation , Immunization , Immunoglobulin Idiotypes/immunology , Multiple Myeloma/immunology , Myeloma Proteins/immunology , Adult , Antigen Presentation , B-Lymphocytes , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Feasibility Studies , Female , Humans , K562 Cells , Lymphocyte Activation , Lymphokines/metabolism , Male , Middle Aged , Multiple Myeloma/therapy
20.
Leuk Lymphoma ; 39(1-2): 165-72, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10975396

ABSTRACT

The majority of follicular lymphoma patients carry a t(14,18) juxtaposing the BCL2 oncogene to the immunoglobulin heavy chain joining region (IgH). Molecular analysis for follicular lymphoma-specific DNA translocations may permit evaluation of minimal residual disease (MRD). We identify extracellular BCL2/IGH transgene DNA in the serum of patients with follicular lymphoma, and evaluate its utility as a surrogate marker. DNA was harvested from both the sera and bone marrow of 5 stage IV follicular lymphoma patients prior to and after chemotherapy and following a novel vaccine-based regimen. Serial PCR amplifications were performed using heminested BCL2-specific major breakpoint cluster region (MBR) primers and the immunoglobulin heavy chain consensus primer. Amplification products were detected by agarose gel electrophoresis, and comparison was made to amplification products from the original tumor biopsy. Results show that four of the five lymphoma patients carried extracellular BCL2/IGH transgene DNA in their serum. The remaining patient did not have an amplification product from either the tumor or the serum, suggesting either the absence of a translocation or the presence of a variant translocation not detectable with this primer set. Transgene DNA was detectable in serum even in patients with MRD, comparing favorably with bone marrow results. In at least one patient, the presence of the transgene in serum at the conclusion of therapy preceded relapse. In conclusion, it seems that tumor-specific, extracellular DNA is present in the serum of follicular lymphoma patients, including those with MRD. Because extracellular DNA may be released into the bloodstream by tumor throughout the body it may be less subject to sampling error, and appears to be an ideal surrogate marker.


Subject(s)
DNA, Neoplasm/blood , Genes, bcl-2/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Joining Region/genetics , Lymphoma, Follicular/genetics , Adult , Aged , Antineoplastic Agents/pharmacology , Biomarkers/blood , Bone Marrow/chemistry , DNA, Neoplasm/drug effects , Female , Humans , Immunoglobulin Heavy Chains/blood , Immunoglobulin Joining Region/blood , Immunotherapy, Active , Lymphoma, Follicular/diagnosis , Male , Middle Aged , Neoplasm, Residual/blood , Neoplasm, Residual/diagnosis , Transgenes , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...