Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 47
1.
Curr Probl Cancer ; 44(6): 100583, 2020 12.
Article En | MEDLINE | ID: mdl-32446637

BACKGROUND: There is increasing use of Phase I statistical designs to find a dose that causes rapidly emerging and particularly concerning severe or life-threatening toxicities (dose-limiting toxicities, DLTs) in a specified percent of patients most commonly 25%. While a convenient statistical framework, the foundation for selecting any specified target DLT rate, and its relevance to the recommended Phase II dose is generally lacking. METHOD: We surveyed 78 medical oncologists, most (69%) with experience as a principal investigator on a Phase I study, to ascertain their opinions related to this approach to Phase I studies and the targets often chosen. RESULTS: Eighty-seven percent of respondents preferred severe toxicities in only 5%-10% of patients, consistent with 58% of respondents noting that 10% or fewer patients experience severe toxicities in the first cycle with standard outpatient treatments. The survey also documented in an example that the majority (62%) of physicians modify their patient selection during the conduct of the study based on observed toxicity and 78% note that higher toxicity is acceptable in patients where a cure is more likely. CONCLUSION: DLT-target rate designs search for a single target that is rarely well-supported in a patient population that is not stable. The most common target used is inconsistent with the toxicity of most clinically used drugs and investigator preference and can lead to the pursuit of unacceptable doses. Use of Phase I trial designs with a target DLT rate should be limited to settings with a well-justified target and should specify how the target relates to the recommended Phase II dose.


Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Clinical Trials, Phase I as Topic/standards , Drug Dosage Calculations , Neoplasms/drug therapy , Oncologists/statistics & numerical data , Research Design/standards , Dose-Response Relationship, Drug , Humans , Neoplasms/pathology , Patient Selection , Surveys and Questionnaires
2.
Oncologist ; 25(5): 382-e776, 2020 05.
Article En | MEDLINE | ID: mdl-31857446

LESSONS LEARNED: Radioembolization with yttrium-90 resin microspheres can be combined safely with full doses of durvalumab and tremelimumab in patients with metastatic colorectal cancer. Regional radioembolization with yttrium-90 resin microspheres did not result in any hepatic or extrahepatic responses to a combination of durvalumab and tremelimumab. The lack of immunomodulatory responses to yttrium-90 on biopsies before and after treatment rules out a potential role for this strategy in converting a "cold tumor" into an "inflamed," immune responsive tumor. BACKGROUND: PD-1 inhibitors have been ineffective in microsatellite stable (MSS) metastatic colorectal cancer (CRC). Preclinical models suggest that radiation therapy may sensitize MSS CRC to PD-1 blockade. METHODS: Patients with MSS metastatic CRC with liver-predominant disease who progressed following at least one prior line of treatment were treated with yttrium-90 (Y90) radioembolization to the liver (SIR-Spheres; Sirtex, Woburn, MA) followed 2-3 weeks later by the combination of durvalumab and tremelimumab. A Simon two-stage design was implemented, with a planned expansion to 18 patients if at least one response was noted in the first nine patients. RESULTS: Nine patients enrolled in the first stage of the study, all with progressive disease (PD) during or after their first two cycles of treatment. Per preplanned design, the study was closed because of futility. No treatment-related grade 3 or greater toxicities were recorded. Correlative studies with tumor biopsies showed low levels of tumor-infiltrating lymphocyte (TIL) infiltration in tumor cancer islands before and after Y90 radioembolization. CONCLUSION: Y90 radioembolization can be added safely to durvalumab and tremelimumab but did not promote tumor-directed immune responses against liver-metastasized MSS CRC.


Colorectal Neoplasms , Embolization, Therapeutic , Liver Neoplasms , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Colorectal Neoplasms/drug therapy , Feasibility Studies , Humans , Liver Neoplasms/drug therapy , Microsatellite Repeats , Treatment Outcome , Yttrium Radioisotopes
3.
Cancer ; 123(23): 4566-4573, 2017 Dec 01.
Article En | MEDLINE | ID: mdl-28832978

BACKGROUND: Targeting the vascular endothelial growth factor (VEGF) pathway has improved outcomes in metastatic renal cell carcinoma (RCC); however, resistance inevitably occurs. CD105 (endoglin) is an angiogenic pathway that is strongly upregulated after VEGF inhibition, potentially contributing to resistance. The authors tested whether TRC105, a monoclonal antibody against endoglin, impacted disease control in patients with previously treated RCC who were receiving bevacizumab. METHODS: Eligible patients with metastatic RCC who had previously received 1 to 4 prior lines of therapy, including VEGF-targeted agents, were randomized 1:1 to receive bevacizumab 10 mg/kg intravenously every 2 weeks (arm A) or the same plus TRC105 10 mg/kg intravenously every 2 weeks (arm B). The primary endpoint was progression-free survival (PFS) at 12 and 24 weeks. Correlative studies included serum transforming growth factor ß (TGFß) and CD105 levels as well as tissue immunostaining for TGFß receptors. RESULTS: Fifty-nine patients were enrolled (28 on arm A and 31 on arm B), and 1 patient on each arm had a confirmed partial response. The median PFS for bevacizumab alone was 4.6 months compared with 2.8 for bevacizumab plus TRC105 (P = .09). Grade ≥ 3 toxicities occurred in 16 patients (57%) who received bevacizumab compared with 19 (61%) who received bevacizumab plus TRC105 (P = .9). Baseline serum TGFß levels below the median (<10.6 ng/mL) were associated with longer median PFS (5.6 vs 2.1 months; P = .014). CONCLUSIONS: TRC105 failed to improve PFS when added to bevacizumab. TGFß warrants further study as a biomarker in RCC. Cancer 2017;123:4566-4573. © 2017 American Cancer Society.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Papillary/drug therapy , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Bevacizumab/administration & dosage , Carcinoma, Papillary/secondary , Carcinoma, Renal Cell/secondary , Female , Follow-Up Studies , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Survival Rate
4.
Blood ; 129(1): 114-125, 2017 01 05.
Article En | MEDLINE | ID: mdl-27760761

Attenuated poxvirus modified vaccinia Ankara (MVA) is a useful viral-based vaccine for clinical investigation, because of its excellent safety profile and property of inducing potent immune responses against recombinant (r) antigens. We developed Triplex by constructing an rMVA encoding 3 immunodominant cytomegalovirus (CMV) antigens, which stimulates a host antiviral response: UL83 (pp65), UL123 (IE1-exon4), and UL122 (IE2-exon5). We completed the first clinical evaluation of the Triplex vaccine in 24 healthy adults, with or without immunity to CMV and vaccinia virus (previous DryVax smallpox vaccination). Three escalating dose levels (DL) were administered IM in 8 subjects/DL, with an identical booster injection 28 days later and 1-year follow-up. Vaccinations at all DL were safe with no dose-limiting toxicities. No vaccine-related serious adverse events were documented. Local and systemic reactogenicity was transient and self-limiting. Robust, functional, and durable Triplex-driven expansions of CMV-specific T cells were detected by measuring T-cell surface levels of 4-1BB (CD137), binding to CMV-specific HLA multimers, and interferon-γ production. Marked and durable CMV-specific T-cell responses were also detected in Triplex-vaccinated CMV-seronegatives, and in DryVax-vaccinated subjects. Long-lived memory effector phenotype, associated with viral control during CMV primary infection, was predominantly found on the membrane of CMV-specific and functional T cells, whereas off-target vaccine responses activating memory T cells from the related herpesvirus Epstein-Barr virus remained undetectable. Combined safety and immunogenicity results of MVA in allogeneic hematopoietic stem cell transplant (HCT) recipients and Triplex in healthy adults motivated the initiation of a placebo-controlled multicenter trial of Triplex in HCT patients. This trial was registered at www.clinicaltrials.gov as #NCT02506933.


Antigens, Viral/immunology , Cytomegalovirus Vaccines/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adult , Cytomegalovirus , Cytomegalovirus Vaccines/adverse effects , Female , Humans , Immediate-Early Proteins/immunology , Male , Middle Aged , Phosphoproteins/immunology , Trans-Activators/immunology , Vaccines, DNA , Viral Matrix Proteins/immunology , Viral Vaccines , Young Adult
5.
Transplantation ; 100(1): 210-6, 2016 Jan.
Article En | MEDLINE | ID: mdl-26680375

BACKGROUND: Immune measurements that distinguish solid organ transplantation (SOT) recipients who control cytomegalovirus (CMV) infection from those who progress to CMV-disease (CMV-dz) may be clinically useful in guiding tailored prevention strategies. We previously reported that elevated plasma levels of the immune-modulator IL-10 are associated with late CMV-dz. Here we evaluate whether IL-10 levels measured soon after prophylaxis discontinuation are predictive of CMV-dz risk. METHODS: Plasma IL-10 levels were quantitatively measured by ELISA kit in 40 D/R SOT patients. All 40 D/R high-risk patients were prospectively followed for at least 12 months post-SOT: 13 subjects developed CMV-dz, all within 6 months of prophylaxis discontinuation. RESULTS: IL-10 was detectable at the first post-prophylaxis measurement for 11 of 13 subjects who developed CMV-dz. In contrast, IL-10 was detectable in only 6 of 27 CMV asymptomatic patients. Monitoring IL-10 plasma levels within 1 month prophylaxis suspension appeared to have clinically useful level of 85% sensitivity and 78% specificity. CONCLUSIONS: The exact role of IL-10 with its multiple immunoregulatory effects during CMV infection is not clear. Moreover, IL-10 production can be influenced by pathological and infectious contexts, and/or anti-rejection immunosuppressant therapy. Despite mechanisms of IL-10 dysregulation may substantially differ among SOT patients, our findings suggest that measurable plasma IL-10 soon after prophylaxis discontinuation may be an adequate indicator of subsequent CMV-dz. If a similar prognostic performance is confirmed in a larger D/R cohort, IL-10 plasma levels could be used to guide the length of prophylaxis, providing a clinically useful means to reduce the incidence of CMV-dz in high risk patients.


Antiviral Agents/administration & dosage , Cytomegalovirus Infections/prevention & control , Drug Monitoring/methods , Ganciclovir/analogs & derivatives , Interleukin-10/blood , Kidney Transplantation/adverse effects , Liver Transplantation/adverse effects , Transplant Recipients , Biomarkers/blood , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/immunology , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay , Ganciclovir/administration & dosage , Humans , Predictive Value of Tests , Prospective Studies , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , Up-Regulation , Valganciclovir
6.
Clin Cancer Res ; 21(19): 4321-6, 2015 Oct 01.
Article En | MEDLINE | ID: mdl-26106072

PURPOSE: Preclinical modeling in non-small cell lung cancer (NSCLC) showed that stimulation with hepatocyte growth factor (HGF), the ligand for MET, could reverse the cytostatic and cytotoxic effects of the EGFR inhibitor erlotinib in erlotinib-sensitive cell lines. Inhibitors of AKT signaling mitigated this HGF-mediated resistance, partially restoring erlotinib activity. We conducted a phase II trial of erlotinib plus MK-2206, a highly selective inhibitor of AKT, in NSCLC patients. EXPERIMENTAL DESIGN: Eligible patients must have progressed following prior benefit from erlotinib, defined as response or stable disease > 12 weeks. Treatment consisted of erlotinib 150 mg orally every day + MK-2206 45 mg orally every alternate day on a 28-day cycle. Primary endpoints were RECIST response rate > 30% (stratum 1: EGFR mutant) and disease control rate (DCR) > 20% at 12 weeks (stratum 2: EGFR wild-type). RESULTS: Eighty patients were enrolled, 45 and 35 in stratum 1 and 2, respectively. Most common attributable adverse events (all grade 3) were rash, diarrhea, fatigue, and mucositis. Response and DCR were, respectively, 9% and 40% in stratum 1; 3% and 47% in stratum 2. Median progression-free survival was 4.4 months in stratum 1 and 4.6 months in stratum 2. CONCLUSIONS: Combination MK-2206 and erlotinib met its primary endpoint in erlotinib-pretreated patients with EGFR wild-type NSCLC. Although activity was seen in EGFR-mutated NSCLC, this did not exceed a priori estimates. AKT pathway inhibition merits further clinical evaluation in EGFR wild-type NSCLC.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/administration & dosage , Female , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Retreatment , Treatment Outcome
7.
PLoS Pathog ; 10(11): e1004524, 2014 Nov.
Article En | MEDLINE | ID: mdl-25412505

Human Cytomegalovirus (HCMV) utilizes two different pathways for host cell entry. HCMV entry into fibroblasts requires glycoproteins gB and gH/gL, whereas HCMV entry into epithelial and endothelial cells (EC) requires an additional complex composed of gH, gL, UL128, UL130, and UL131A, referred to as the gH/gL-pentamer complex (gH/gL-PC). While there are no established correlates of protection against HCMV, antibodies are thought to be important in controlling infection. Neutralizing antibodies (NAb) that prevent gH/gL-PC mediated entry into EC are candidates to be assessed for in vivo protective function. However, these potent NAb are predominantly directed against conformational epitopes derived from the assembled gH/gL-PC. To address these concerns, we constructed Modified Vaccinia Ankara (MVA) viruses co-expressing all five gH/gL-PC subunits (MVA-gH/gL-PC), subsets of gH/gL-PC subunits (gH/gL or UL128/UL130/UL131A), or the gB subunit from HCMV strain TB40/E. We provide evidence for cell surface expression and assembly of complexes expressing full-length gH or gB, or their secretion when the corresponding transmembrane domains are deleted. Mice or rhesus macaques (RM) were vaccinated three times with MVA recombinants and serum NAb titers that prevented 50% infection of human EC or fibroblasts by HCMV TB40/E were determined. NAb responses induced by MVA-gH/gL-PC blocked HCMV infection of EC with potencies that were two orders of magnitude greater than those induced by MVA expressing gH/gL, UL128-UL131A, or gB. In addition, MVA-gH/gL-PC induced NAb responses that were durable and efficacious to prevent HCMV infection of Hofbauer macrophages, a fetal-derived cell localized within the placenta. NAb were also detectable in saliva of vaccinated RM and reached serum peak levels comparable to NAb titers found in HCMV hyperimmune globulins. This vaccine based on a translational poxvirus platform co-delivers all five HCMV gH/gL-PC subunits to achieve robust humoral responses that neutralize HCMV infection of EC, placental macrophages and fibroblasts, properties of potential value in a prophylactic vaccine.


Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cytomegalovirus Infections , Cytomegalovirus Vaccines , Cytomegalovirus , Multiprotein Complexes , Viral Envelope Proteins , Animals , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/genetics , Cytomegalovirus Vaccines/immunology , Female , Humans , Macaca mulatta , Mice , Mice, Inbred BALB C , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
8.
J Virol ; 87(3): 1322-32, 2013 Feb.
Article En | MEDLINE | ID: mdl-23152525

Neutralizing antibodies (NAb) are important for interfering with horizontal transmission of human cytomegalovirus (HCMV) leading to primary and congenital HCMV infection. Recent findings have shown that a pentameric virion complex formed by the glycoproteins gH/gL, UL128, UL130, and UL131A (UL128C) is required for HCMV entry into epithelial/endothelial cells (Epi/EC) and is the target of potent NAb in HCMV-seropositive individuals. Using bacterial artificial chromosome technology, we have generated a modified vaccinia Ankara virus (MVA) that stably coexpresses all 5 rhesus CMV (RhCMV) proteins homologous to HCMV UL128C, termed MVA-RhUL128C. Coimmunoprecipitation confirmed the interaction of RhgH with the other 4 RhCMV subunits of the pentameric complex. All 8 RhCMV-naïve rhesus macaques (RM) vaccinated with MVA-RhUL128C developed NAb that blocked infection of monkey kidney epithelial cells (MKE) and rhesus fibroblasts. NAb titers induced by MVA-RhUL128C measured on both cell types at 2 to 6 weeks postvaccination were comparable to levels observed in naturally infected RM. In contrast, MVA expressing a subset of RhUL128C proteins or RhgB glycoprotein only minimally stimulated NAb that inhibited infection of MKE. In addition, following subcutaneous RhCMV challenge at 8 weeks postvaccination, animals vaccinated with MVA-RhUL128C showed reduced plasma viral loads. These results indicate that MVA expressing the RhUL128C induces NAb inhibiting RhCMV entry into both Epi/EC and fibroblasts and limits RhCMV replication in RM. This novel approach is the first step in developing a prophylactic HCMV vaccine designed to interfere with virus entry into major cell types permissive for viral replication, a required property of an effective vaccine.


Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cytomegalovirus Vaccines/immunology , Viral Envelope Proteins/immunology , Animals , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/administration & dosage , DNA, Viral/chemistry , DNA, Viral/genetics , Drug Carriers , Epithelial Cells/virology , Fibroblasts/virology , Genetic Vectors , Macaca mulatta , Molecular Sequence Data , Sequence Analysis, DNA , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Viral Envelope Proteins/administration & dosage , Viremia/prevention & control , Virus Internalization
9.
J Infect Dis ; 205(8): 1294-304, 2012 Apr 15.
Article En | MEDLINE | ID: mdl-22402037

BACKGROUND: It has been reported that cytomegalovirus (CMV) pp65-specific T cells can protect hematopoietic cell transplant (HCT) recipients from CMV complications. Two candidate CMV peptide vaccines composed of the HLA A*0201 pp65(495-503) cytotoxic CD8(+) T-cell epitope fused to 2 different universal T-helper epitopes (either the synthetic Pan DR epitope [PADRE] or a natural Tetanus sequence) were clinically evaluated for safety and ability to elicit pp65 T cells in HLA A*0201 healthy volunteers. METHODS: Escalating doses (0.5, 2.5, 10 mg) of PADRE or Tetanus pp65(495-503) vaccines with (30 adults) or without (28 adults) PF03512676 adjuvant were administered by subcutaneous injection every 3 weeks for a total of 4 injections. RESULTS: No serious adverse events were reported, although vaccines used in combination with PF03512676 had enhanced reactogenicity. Ex vivo responses were detected by flow cytometry exclusively in volunteers who received the vaccine coadministered with PF03512676. In addition, using a sensitive in vitro stimulation system, vaccine-elicited pp65(495-503) T cells were expanded in 30% of volunteers injected solely with the CMV peptides and in all tested subjects receiving the vaccines coinjected with PF03512676. CONCLUSIONS: Acceptable safety profiles and vaccine-driven expansion of pp65(495-503) T cells in healthy adults support further evaluation of CMV peptide vaccines combined with PF03512676 in the HCT setting. CLINICAL TRIALS REGISTRATION: NCT00722839.


Adjuvants, Immunologic/administration & dosage , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Vaccines/immunology , Malaria Vaccines/immunology , Oligodeoxyribonucleotides/administration & dosage , Tetanus Toxoid/immunology , Adjuvants, Immunologic/adverse effects , Adolescent , Adult , Amino Acid Sequence , CD8-Positive T-Lymphocytes/physiology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/adverse effects , Dose-Response Relationship, Immunologic , Epitopes , Female , Humans , Male , Middle Aged , Oligodeoxyribonucleotides/adverse effects , Recombinant Proteins/immunology , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/adverse effects , Vaccines, Synthetic , Young Adult
10.
Cancer Chemother Pharmacol ; 69(3): 835-43, 2012 Mar.
Article En | MEDLINE | ID: mdl-22105720

BACKGROUND: 3-Aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) is a novel small-molecule ribonucleotide reductase inhibitor. This study was designed to estimate the maximum tolerated dose (MTD) and oral bioavailability of 3-AP in patients with advanced-stage solid tumors. METHODS: Twenty patients received one dose of intravenous and subsequent cycles of oral 3-AP following a 3 + 3 patient dose escalation. Intravenous 3-AP was administered to every patient at a fixed dose of 100 mg over a 2-h infusion 1 week prior to the first oral cycle. Oral 3-AP was administered every 12 h for 5 consecutive doses on days 1-3, days 8-10, and days 15-17 of every 28-day cycle. 3-AP was started at 50 mg with a planned dose escalation to 100, 150, and 200 mg. Dose-limiting toxicities (DLT) and bioavailability were evaluated. RESULTS: Twenty patients were enrolled. For dose level 1 (50 mg), the second of three treated patients had a DLT of grade 3 hypertension. In the dose level 1 expansion cohort, three patients had no DLTs. No further DLTs were encountered during escalation until the 200-mg dose was reached. At the 200 mg 3-AP dose level, two treated patients had DLTs of grade 3 hypoxia. One additional DLT of grade 4 febrile neutropenia was subsequently observed at the de-escalated 150 mg dose. One DLT in 6 evaluable patients established the MTD as 150 mg per dose on this dosing schedule. Responses in the form of stable disease occurred in 5 (25%) of 20 patients. The oral bioavailability of 3-AP was 67 ± 29% and was consistent with the finding that the MTD by the oral route was 33% higher than by the intravenous route. CONCLUSIONS: Oral 3-AP is well tolerated and has an MTD similar to its intravenous form after accounting for the oral bioavailability. Oral 3-AP is associated with a modest clinical benefit rate of 25% in our treated patient population with advanced solid tumors.


Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Pyridines/pharmacokinetics , Thiosemicarbazones/pharmacokinetics , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Biological Availability , California , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Injections, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/pathology , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/therapeutic use , Thiosemicarbazones/administration & dosage , Thiosemicarbazones/adverse effects , Thiosemicarbazones/therapeutic use , Treatment Outcome
11.
Blood ; 118(8): 2159-69, 2011 Aug 25.
Article En | MEDLINE | ID: mdl-21719601

The serine proteases, neutrophil elastase (HNE) and proteinase 3 (PR3), are aberrantly expressed in human myeloid leukemias. T-cell responses to these proteins have been correlated with remission in patients with chronic myeloid leukemia (CML). Human PR3/HNE-specific CD8(+) T cells predominantly recognize a nonameric HLA-A2-restricted T-cell epitope called PR1 which is conserved in both Ags. However, CML patients have CD8(+) T cells in peripheral blood recognizing an additional HLA-A2 epitope termed PR2. To assess immunologic properties of these Ags, novel recombinant vaccinia viruses (rVV) expressing PR3 and HNE were evaluated in HLA-A2 transgenic (Tg) mice (HHDII). Immunization of HHDII mice with rVV-PR3 elicited a robust PR3-specific CD8(+) T-cell response dominated by recognition of PR2, with minimal recognition of the PR1 epitope. This result was unexpected, because the PR2 peptide has been reported to bind poorly to HLA. To account for these findings, we proposed that HHDII mice negatively selected PR1-specific T cells because of the presence of this epitope within murine PR3 and HNE, leading to immunodominance of PR2-specific responses. PR2-specific splenocytes are cytotoxic to targets expressing naturally processed PR3, though PR1-specific splenocytes are not. We conclude that PR2 represents a functional T-cell epitope recognized in mice and human leukemia patients. These studies are registered at www.clinicaltrials.gov as NCT00716911.


Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Amino Acid Sequence , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cytoplasmic Granules/enzymology , Cytoplasmic Granules/immunology , Epitopes, T-Lymphocyte/genetics , HLA-A2 Antigen/genetics , HLA-A2 Antigen/metabolism , Hematopoietic Stem Cell Transplantation , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukocyte Elastase/genetics , Leukocyte Elastase/immunology , Mice , Mice, Transgenic , Molecular Sequence Data , Myeloblastin/genetics , Myeloblastin/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Homology, Amino Acid , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology
12.
Transpl Int ; 24(9): 920-31, 2011 Sep.
Article En | MEDLINE | ID: mdl-21672050

Antiviral prophylaxis has proved successful for prevention of cytomegalovirus (CMV) disease in solid organ transplant (SOT) patients; though emerging data suggest that antiviral agents interfere with immunity, and may inhibit immune priming. In this context, we investigated levels and phenotype of primary CMV-specific immune responses that developed during antiviral prophylaxis in a cohort of CMV seronegative recipients (R(-) ) of a SOT from a seropositive donor (D(+) ). We longitudinally monitored CMV viral load, antibodies and levels of the negative immuno-modulator IL-10. PBMC were stimulated with CMV-specific peptide libraries to measure CD137 activation marker on CMV-specific T-cells and levels of PD-1 receptor, which is over expressed on exhausted T-cells. Unexpectedly, the majority (13/18) of D(+) R(-) patients who developed a primary CMV response showed early post-transplant CMV-specific responses, though levels of PD-1 on CMV-specific T-cells remained elevated throughout prophylaxis. A strong inverse association was found between levels of plasma IL-10 and CMV-specific cellular immune responses. Our study suggests that during prophylaxis, subclinical CMV infection might have occurred in the D(+) R(-) patients, and primary CMV-specific responses were detected early post-transplant when levels of plasma IL-10 were low. Extended prophylaxis or antiviral treatment did not appear to suppress CMV-specific antibodies or T-cells, which, however, showed exhaustion phenotypes.


Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Cytomegalovirus/immunology , Ganciclovir/analogs & derivatives , Kidney Transplantation/methods , Liver Transplantation/methods , Adult , Aged , Antigens, Viral/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Female , Ganciclovir/therapeutic use , Humans , Immediate-Early Proteins/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Male , Middle Aged , Phosphoproteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/virology , Valganciclovir , Viral Load , Viral Matrix Proteins/immunology
13.
Int J Clin Oncol ; 16(5): 494-9, 2011 Oct.
Article En | MEDLINE | ID: mdl-21431345

BACKGROUND: This study was conducted to evaluate the efficacy of the epidermal growth factor receptor tyrosine kinase inhibitor gefitinib with pegylated-IFNα (PEG-IFNα) in patients with advanced renal cell carcinoma. METHODS: Progression-free survival (PFS) rate at 6 months >50% was considered promising for further evaluation. Patients with unresectable or metastatic disease, unlimited prior therapies, and adequate performance status and end-organ function were eligible. PEG-IFNα was dosed subcutaneously once weekly (initially 6 µg/kg/week, later reduced to 4 µg/kg/week) for 12 weeks. Gefitinib was given 250 mg orally once daily until progression or intolerance. RESULTS: Twenty-one patients were accrued. Fourteen patients had a prior nephrectomy, and twelve had prior systemic therapy. The 6-month PFS was 29% (95%CI 15-56%). Best responses by RECIST criteria: complete, partial (1, plus 3 unconfirmed) stable (Uhlman et al. Clin Cancer Res 1:913-920, 1995), and progression (Sirotnak et al. Clin Cancer Res 6:4885-4892, 2000). Response duration: complete response (35+ months) and partial response (2, 3, 3, 37 months). Median PFS and overall survival were 5.3 (95%CI 3-10.1) and 13.6 (95%CI 10.3-NA) months, respectively. Most common toxicities included myelosuppression, rash, and nausea. CONCLUSIONS: Although generally well tolerated, gefitinib plus PEG-IFNα did not meet the pre-specified 6-month PFS rate >50%. Further evaluation of similar regimens would require appropriate molecular selection of subjects most likely to benefit. Thus, preclinical studies to determine candidate predictive markers for this combination are warranted.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Renal Cell/drug therapy , Interferon-alpha/administration & dosage , Kidney Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Quinazolines/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Renal Cell/pathology , Disease-Free Survival , Female , Gefitinib , Humans , Interferon-alpha/adverse effects , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Polyethylene Glycols/adverse effects , Quinazolines/adverse effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects
14.
Clin Lung Cancer ; 12(1): 33-7, 2011 Jan.
Article En | MEDLINE | ID: mdl-21273177

BACKGROUND: The proteasome inhibitor bortezomib sensitizes tumor cells to chemotherapy-induced apoptosis. In preclinical non-small-cell lung cancer (NSCLC) models, p53-dependent growth arrest after bortezomib treatment resulted in reduced cytotoxicity if bortezomib preceded docetaxel. The reverse sequence of docetaxel before bortezomib was associated with increased apoptosis, cleavage of caspase-3 and PARP (poly [ADP-ribose] polymerase), and reduction in Bcl-2. A prospective randomized phase II trial of concurrent versus sequential docetaxel and bortezomib was conducted to assess whether administration sequence resulted in measurable clinical differences. PATIENTS AND METHODS: Previously treated patients with advanced NSCLC were randomized to concurrent (CON) or sequential (SEQ) docetaxel (75 mg/m² intravenous [I.V.]) followed by bortezomib, every 3 weeks. In the CON arm, bortezomib (1.6 mg/m² I.V.) was given on days 1 and 8, and in the SEQ arm, it was given on days 2 and 8. Previous erlotinib as well as treated or controlled brain metastases were allowed. The primary endpoint was objective response rate (RR); progression-free (PFS) and overall survival (OS) were secondary endpoints. RESULTS: Eighty-one patients were randomized (40 CON and 41 SEQ). Grade 3+ toxicities were mostly due to myelosuppression. One patient each had grade 4 hyponatremia and syncope. Toxicities were similar between the arms. There was 1 treatment-related death in the SEQ arm. There were 8 partial responders, 4 in each arm, for an overall RR of 10%. Disease control rate was similar in both arms (50% vs. 49%). Median PFS was 12 weeks in the CON arm and 11 weeks in the SEQ arm. Median OS times in the CON and SEQ arms were 13.3 and 10.5 months, respectively. CONCLUSION: Docetaxel plus bortezomib given sequentially or concurrently has similar RR and PFS. Median survival in the SEQ arm exceeds published survival estimates for either agent alone or in combination. Any further studies in this population would require molecular characterization of a phenotype most likely to benefit from proteasome inhibitor therapy.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Pyrazines/therapeutic use , Taxoids/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Bortezomib , California , Carcinoma, Non-Small-Cell Lung/mortality , Disease-Free Survival , Docetaxel , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Middle Aged , Pyrazines/administration & dosage , Pyrazines/adverse effects , Taxoids/administration & dosage , Taxoids/adverse effects
15.
Neurosci Lett ; 486(3): 136-40, 2010 Dec 17.
Article En | MEDLINE | ID: mdl-20850505

In a large Scottish pedigree, a balanced translocation t (1;11)(q42.1;q14.3) disrupting the DISC1 and DISC2 genes segregates with major mental illness, including schizophrenia and depression. A frame-shift carboxyl-terminal deletion was reported in DISC1 in an American family with schizophrenia, but subsequently found in two controls. Herein, we test one hypothesis utilizing a large scale case-control mutation analysis: uncommon DISC1 variants are associated with high risk for bipolar spectrum disorder. We have analyzed the regions of likely functional significance in the DISC1 gene in 504 patients with bipolar spectrum disorder and 576 ethnically similar controls. Five patients were heterozygous for ultra-rare protein structural variants not found in the 576 controls (p=0.02, one-sided Fisher's exact test) and shown to be ultra-rare by their absence in a pool of 10,000 control alleles. In our sample, ultra-rare (private) protein structural variants in DISC1 are associated with an estimated attributable risk of about 0.5% in bipolar spectrum disorder. These data are consistent with: (i) the high frequency of depression in the large Scottish family with a translocation disrupting DISC1; (ii) linkage disequilibrium analysis demonstrating haplotypes associated with relatively small increases in risk for bipolar disorder (<3-fold odds ratio). The data illustrate how low/moderate risk haplotypes that might be found by the HapMap project can be followed up by resequencing to identify protein structural variants with high risk, low frequency and of potential clinical utility.


Bipolar Disorder/diagnosis , Bipolar Disorder/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Nerve Tissue Proteins/genetics , Adult , Bipolar Disorder/ethnology , Case-Control Studies , Cohort Studies , Female , Humans , Linkage Disequilibrium/genetics , Male , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Risk Assessment/methods , Scotland/ethnology , Translocation, Genetic/genetics
16.
PLoS One ; 4(7): e6121, 2009 Jul 01.
Article En | MEDLINE | ID: mdl-19568434

BACKGROUND: Schizophrenia is a severe disabling brain disease affecting about 1% of the population. Individual microRNAs (miRNAs) affect moderate downregulation of gene expression. In addition, components required for miRNA processing and/or function have also been implicated in X-linked mental retardation, neurological and neoplastic diseases, pointing to the wide ranging involvement of miRNAs in disease. METHODS AND FINDINGS: To explore the role of miRNAs in schizophrenia, 59 microRNA genes on the X-chromosome were amplified and sequenced in males with (193) and without (191) schizophrenia spectrum disorders to test the hypothesis that ultra-rare mutations in microRNA collectively contribute to the risk of schizophrenia. Here we provide the first association of microRNA gene dysfunction with schizophrenia. Eight ultra-rare variants in the precursor or mature miRNA were identified in eight distinct miRNA genes in 4% of analyzed males with schizophrenia. One ultra-rare variant was identified in a control sample (with a history of depression) (8/193 versus 1/191, p = 0.02 by one-sided Fisher's exact test, odds ratio = 8.2). These variants were not found in an additional 7,197 control X-chromosomes. CONCLUSIONS: Functional analyses of ectopically expressed copies of the variant miRNA precursors demonstrate loss of function, gain of function or altered expression levels. While confirmation is required, this study suggests that microRNA mutations can contribute to schizophrenia.


Genetic Diseases, X-Linked , MicroRNAs/genetics , Schizophrenia/genetics , Base Sequence , Blotting, Northern , Cell Line , Humans , Male , Plasmids , Sequence Homology, Nucleic Acid
17.
Anticancer Drugs ; 20(3): 179-84, 2009 Mar.
Article En | MEDLINE | ID: mdl-19396016

Prostate cancer cells undergo neuroendocrine differentiation during androgen deprivation and secrete neuropeptides, hence activating androgen receptor-regulated genes. Src-family protein kinases are involved in neuropeptide-induced prostate cancer growth and migration. A phase II trial of AZD0530, an oral Src-family kinase inhibitor, in patients with advanced castration resistant prostate cancer was conducted. The primary endpoint was prostate cancer-specific antigen (PSA) response rate, defined as a 30% or greater decrease. A two-stage Simon design was used. Eligibility criteria included documentation of castration resistance (including antiandrogen withdrawal), adequate end-organ function, and performance status, and not more than one prior taxane-based chemotherapy regimen. AZD0530 was given at 175 mg orally once daily continuously. Rapid accrual led to 28 patients registering in the first stage. Median age was 67 years. Sixteen patients had performance status (PS) 0, eight patients had PS 1, and four patients had PS 2. Nine patients (32%) had prior docetaxel-based chemotherapy. Five patients had transient PSA reductions not meeting PSA response criteria. Median progression-free survival time was 8 weeks. Treatment was generally well tolerated. AZD0530, a potent oral Src kinase inhibitor, is feasible and tolerable in this pretreated patient population but possessed little clinical efficacy as monotherapy. Strong preclinical evidence warrants further investigation of AZD0530 in earlier-stage prostate cancer or as combination therapy.


Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Benzodioxoles/therapeutic use , Prostatic Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , src-Family Kinases/antagonists & inhibitors , Adenocarcinoma/blood , Adenocarcinoma/surgery , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Benzodioxoles/adverse effects , Benzodioxoles/pharmacology , Chemical and Drug Induced Liver Injury/etiology , Combined Modality Therapy , Disease-Free Survival , Drug Resistance, Neoplasm , Gastrointestinal Diseases/chemically induced , Humans , Lymphopenia/chemically induced , Male , Middle Aged , Orchiectomy , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/surgery , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/adverse effects , Quinazolines/pharmacology
18.
Blood ; 113(25): 6465-76, 2009 Jun 18.
Article En | MEDLINE | ID: mdl-19369230

Reconstitution of cytomegalovirus (CMV)-specific CD8(+) T cells is essential to the control of CMV infection in CMV-positive recipients (R(+)) after allogeneic hematopoietic stem cell transplantation (HCT). Six-color flow cytometry was used to assess the functional profile of CMV-specific CD8(+) T cells in 62 of 178 R(+) HCT recipients followed virologically for CMV reactivation. R(+) recipients receiving grafts from CMV-negative donors (D(-); D(-)/R(+)) reconstituted fewer multifunctional CD8(+) T cells expressing tumor necrosis factor-alpha (TNF-alpha), macrophage inflammatory protein-1beta (MIP-1beta), and CD107 in addition to interferon-gamma (IFN-gamma), compared with D(+)/R(+) recipients. Unlike monofunctional CD8(+) T cells secreting IFN-gamma, which were abundantly generated during CMV reactivation in D(-)/R(+) recipients, the relative lack of multifunctional CD8(+) T cells persisted until at least 1 year post-HCT. D(-)/R(+) recipients were more likely to require recurrent and prolonged use of antivirals. These findings were robust to statistical adjustment for pretransplant factors, as well as for posttransplant factors including graft-versus-host disease (GVHD) and its treatment by steroids. These analyses suggest that D(+)/R(+) transplants, on average, generate higher levels of multifunctional CMV-specific T cells and require less antiviral therapy compared with D(-)/R(+) HCT recipients. These results highlight the benefit of D(+) donors in improving outcomes of R(+) HCT recipients by reducing the duration and recurrent need of antiviral treatment, aided by increased levels of multifunctional CMV-specific T cells.


Bone Marrow Transplantation/statistics & numerical data , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/isolation & purification , Peripheral Blood Stem Cell Transplantation/statistics & numerical data , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , Tissue Donors , Transplantation, Homologous/statistics & numerical data , Adult , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Bone Marrow Transplantation/adverse effects , CD8-Positive T-Lymphocytes/metabolism , Cohort Studies , Cytokines/metabolism , Cytomegalovirus/physiology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/transmission , Cytomegalovirus Vaccines , Disease Transmission, Infectious , Female , Graft Survival/immunology , Hematologic Neoplasms/blood , Hematologic Neoplasms/immunology , Hematologic Neoplasms/surgery , Humans , Immunity, Cellular , Male , Middle Aged , Peripheral Blood Stem Cell Transplantation/adverse effects , Phosphoproteins/immunology , Siblings , Transplantation, Homologous/adverse effects , Treatment Outcome , Viral Matrix Proteins/immunology , Virus Activation
19.
Cancer Chemother Pharmacol ; 64(4): 777-83, 2009 Sep.
Article En | MEDLINE | ID: mdl-19169683

PURPOSE: To evaluate the response to lapatinib, an inhibitor of epidermal growth factor receptors 1 and 2, in patients with advanced bilary tree cancer (BTC) and hepatocellular cancer (HCC). METHODS: Lapatinib was dosed at 1,500 mg/day orally continuously. RESULTS: Fifty-seven patients were accrued (BTC 17, HCC 40). Therapy was well tolerated. The response in BTC was 0% and in HCC was 5%. The progression free survival (PFS) for BTC and HCC patients was 1.8 (95% CI: 1.7-5.2) months and 2.3 (95% CI: 1.7-5.6) months. The median survival for BTC and HCC patients was 5.2 (95% CI 3.3-infinity) months and 6.2 (95% CI: 5.1-infinity) months. EGFR genotyping indicated HCC patients with <20 repeats have the lowest PFS. The occurrence of any skin rash significantly prolonged PFS and survival. CONCLUSIONS: Lapatinib was well-tolerated. There was evidence of activity in HCC, but therapy with lapatinib did not meet the predefined efficacy rate.


Antineoplastic Agents/therapeutic use , Biliary Tract Neoplasms/drug therapy , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Quinazolines/therapeutic use , Antineoplastic Agents/adverse effects , Biliary Tract Neoplasms/pathology , Carcinoma, Hepatocellular/pathology , Humans , Lapatinib , Liver Neoplasms/pathology , Quinazolines/adverse effects , Survival Analysis
20.
J Infect Dis ; 197(1): 25-33, 2008 Jan 01.
Article En | MEDLINE | ID: mdl-18171281

Immunological parameters that distinguish solid-organ transplant (SOT) recipients at risk for life-threatening cytomegalovirus (CMV) disease are being actively pursued to aid posttransplant management. A candidate marker is programmed death (PD)-1 receptor, whose overexpression has been associated with disease progression during persistent viral infections. To determine whether levels of this negative regulator of T cell activity are altered in SOT recipients with symptoms of CMV disease, a comparative PD-1 expression analysis was done in healthy, CMV-positive individuals and in liver transplant recipients. PD-1 levels were measured among the total population of CD8(+) and CD8(+) T cells binding to CMV-specific major histocompatibility complex class I tetramers. Minimal PD-1 expression was found in the healthy, CMV-positive cohort, and symptomatic SOT recipients had significantly higher PD-1 levels. PD-1 up-regulation was significantly associated with incipient and overt CMV disease and with viremia. Our findings suggest that PD-1 could be developed as a prognostic tool to predict CMV disease and guide therapeutic interventions.


Antigens, CD/metabolism , Apoptosis Regulatory Proteins/metabolism , CD8-Positive T-Lymphocytes/virology , Cytomegalovirus Infections/physiopathology , Liver Transplantation/adverse effects , Viremia/physiopathology , Adolescent , Adult , Biomarkers/blood , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Humans , Longitudinal Studies , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor , Up-Regulation , Viremia/blood
...