Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
PLoS Genet ; 13(11): e1007087, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29166413

ABSTRACT

Multiple Myeloma (MM) is a plasma cell malignancy with significantly greater incidence and mortality rates among African Americans (AA) compared to Caucasians (CA). The overall goal of this study is to elucidate differences in molecular alterations in MM as a function of self-reported race and genetic ancestry. Our study utilized somatic whole exome, RNA-sequencing, and correlated clinical data from 718 MM patients from the Multiple Myeloma Research Foundation CoMMpass study Interim Analysis 9. Somatic mutational analyses based upon self-reported race corrected for ancestry revealed significant differences in mutation frequency between groups. Of interest, BCL7A, BRWD3, and AUTS2 demonstrate significantly higher mutation frequencies among AA cases. These genes are all involved in translocations in B-cell malignancies. Moreover, we detected a significant difference in mutation frequency of TP53 and IRF4 with frequencies higher among CA cases. Our study provides rationale for interrogating diverse tumor cohorts to best understand tumor genomics across populations.


Subject(s)
Interferon Regulatory Factors/genetics , Microfilament Proteins/genetics , Multiple Myeloma/genetics , Oncogene Proteins/genetics , Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Adult , Black People/genetics , Cytoskeletal Proteins , Exome/genetics , Female , Genotype , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Multiple Myeloma/epidemiology , Multiple Myeloma/pathology , Mutation , Mutation Rate , Racial Groups , White People/genetics
2.
Br J Haematol ; 180(1): 71-81, 2018 01.
Article in English | MEDLINE | ID: mdl-29105742

ABSTRACT

We have developed an automated assay to enumerate and characterize circulating multiple myeloma cells (CMMC) from peripheral blood of patients with plasma cell disorders. CMMC show expression of genes characteristic of myeloma and fluorescence in situ hybridisation results on CMMC correlated well with bone marrow results. We enumerated CMMC from over 1000 patient samples including separate cohorts of newly diagnosed multiple myeloma and high/intermediate risk smouldering multiple myeloma (SMM) with clinical follow-up data. In newly diagnosed myeloma patient samples, CMMC counts correlated with other clinical measures of disease burden, including the percentage of bone marrow plasma cells, serum M protein, and International Staging System stage. CMMC counts decreased significantly from baseline when a remission was achieved due to treatment (P < 0·001). Patients with CMMC counts ≥100 at remission showed reduced survival relative to patients with CMMC counts <100. Patients with undetectable CMMC in remission showed further overall survival benefits. In the SMM cohort, there was a trend toward higher CMMC in patients with higher-risk myeloma precursor states. Significantly higher CMMC counts were observed between intermediate/high risk SMM patients that progressed versus those without progression (P = 0·031). CMMC allow a non-invasive means of monitoring tumour biology and may have use as a prognostic test for patients with plasma cell disorders.


Subject(s)
Cell Count , Multiple Myeloma/diagnosis , Neoplasms, Plasma Cell/diagnosis , Neoplastic Cells, Circulating/pathology , Adult , Aged , Bone Marrow/pathology , Cohort Studies , Diagnosis, Differential , Female , Flow Cytometry/methods , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Neoplasms, Plasma Cell/blood , Neoplasms, Plasma Cell/genetics , Neoplasms, Plasma Cell/mortality , Neoplastic Cells, Circulating/metabolism , Prognosis , ROC Curve , Reproducibility of Results , Sensitivity and Specificity
3.
Vaccines (Basel) ; 11(12)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38140193

ABSTRACT

We previously reported a protective antibody response in mice immunized with synthetic microparticle vaccines made using layer-by-layer fabrication (LbL-MP) and containing the conserved T1BT* epitopes from the P. falciparum circumsporozoite protein. To further optimize the vaccine candidate, a benchtop tangential flow filtration method (LbL-by-TFF) was developed and utilized to produce vaccine candidates that differed in the status of base layer crosslinking, inclusion of a TLR2 ligand in the antigenic peptide, and substitution of serine or alanine for an unpaired cysteine residue in the T* epitope. Studies in mice revealed consistent superiority of the Pam3Cys-modified candidates and a modest benefit of base layer crosslinking, as evidenced by higher and more persistent antibody titers (up to 18 months post-immunization), a qualitative improvement of T-cell responses toward a Th1 phenotype, and greater protection from live parasite challenges compared to the unmodified prototype candidate. Immunogenicity was also tested in a non-human primate model, the rhesus macaque. Base layer-crosslinked LbL-MP loaded with T1BT* peptide with or without covalently linked Pam3Cys elicited T1B-specific antibody responses and T1BT*-specific T-cell responses dominated by IFNγ secretion with lower levels of IL-5 secretion. The Pam3Cys-modified construct was more potent, generating antibody responses that neutralized wild-type P. falciparum in an in vitro hepatocyte invasion assay. IgG purified from individual macaques immunized with Pam3Cys.T1BT* LbL-MP protected naïve mice from challenges with transgenic P. berghei sporozoites that expressed the full-length PfCS protein, with 50-88% of passively immunized mice parasite-free for ≥15 days. Substitution of serine for an unpaired cysteine in the T* region of the T1BT* subunit did not adversely impact immune potency in the mouse while simplifying the manufacture of the antigenic peptide. In a Good Laboratory Practices compliant rabbit toxicology study, the base layer-crosslinked, Pam3Cys-modified, serine-substituted candidate was shown to be safe and immunogenic, eliciting parasite-neutralizing antibody responses and establishing the dose/route/regimen for a clinical evaluation of this novel synthetic microparticle pre-erythrocytic malaria vaccine candidate.

4.
Peptides ; 29(3): 479-86, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17942192

ABSTRACT

Vasoactive intestinal peptide (VIP) binds to two receptors, VPAC1 and VPAC2. Non-selective VIP antagonists have been shown to inhibit human cancer cell proliferation and reduce tumor growth in mice. Many human cancers over-express VPAC1 but not VPAC2. We show that VPAC1-selective antagonists can inhibit human cancer cell proliferation and identify five positions in the VPAC1-selective antagonist PG 97-269 that may be responsible for VPAC1 selectivity. Position 16 appears to be particularly critical for selectivity, as demonstrated in the replacement of Arg16 of PG 97-269 with the native VIP amino acid; this single change results in greatly reduced VPAC1 binding and selectivity. Finally, we show that site-specific conjugation with a 22kDa polyethylene glycol (PEG) at the C-terminus of VPAC1-selective antagonists further improves VPAC1-selective binding and has minimal effect on antagonistic activity. Our studies have further solidified VPAC1 as a cancer target and offer the possibility of generating highly potent VPAC1-selective antagonists with minimal number of mutations to reduce the risk of immunogenicity and potentially prolonged duration of action to allow more efficient treatment regimen.


Subject(s)
Cell Proliferation/drug effects , Peptide Fragments/pharmacology , Polyethylene Glycols/chemistry , Receptors, Vasoactive Intestinal Polypeptide, Type I/antagonists & inhibitors , Amino Acid Sequence , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Electrophoresis, Polyacrylamide Gel , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Binding/drug effects , Radioimmunoassay , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship , Vasoactive Intestinal Peptide/chemistry , Vasoactive Intestinal Peptide/pharmacology
5.
Blood Cancer J ; 8(3): 35, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29563506

ABSTRACT

Multiple myeloma (MM) is a disease of copy number variants (CNVs), chromosomal translocations, and single-nucleotide variants (SNVs). To enable integrative studies across these diverse mutation types, we developed a capture-based sequencing platform to detect their occurrence in 465 genes altered in MM and used it to sequence 95 primary tumor-normal pairs to a mean depth of 104×. We detected cases of hyperdiploidy (23%), deletions of 1p (8%), 6q (21%), 8p (17%), 14q (16%), 16q (22%), and 17p (4%), and amplification of 1q (19%). We also detected IGH and MYC translocations near expected frequencies and non-silent SNVs in NRAS (24%), KRAS (21%), FAM46C (17%), TP53 (9%), DIS3 (9%), and BRAF (3%). We discovered frequent mutations in IGLL5 (18%) that were mutually exclusive of RAS mutations and associated with increased risk of disease progression (p = 0.03), suggesting that IGLL5 may be a stratifying biomarker. We identified novel IGLL5/IGH translocations in two samples. We subjected 15 of the pairs to ultra-deep sequencing (1259×) and found that although depth correlated with number of mutations detected (p = 0.001), depth past ~300× added little. The platform provides cost-effective genomic analysis for research and may be useful in individualizing treatment decisions in clinical settings.


Subject(s)
Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains, Surrogate/genetics , Multiple Myeloma/genetics , Point Mutation , Translocation, Genetic , Biomarkers, Tumor , DNA Copy Number Variations , Gene Expression , Genes, myc , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Membrane Proteins/genetics , Polymorphism, Single Nucleotide , Prognosis
6.
Vaccine ; 31(15): 1898-904, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23481177

ABSTRACT

Epitopes of the circumsporozoite (CS) protein of Plasmodium falciparum, the most pathogenic species of the malaria parasite, have been shown to elicit protective immunity in experimental animals and human volunteers. The mechanisms of immunity include parasite-neutralizing antibodies that can inhibit parasite motility in the skin at the site of infection and in the bloodstream during transit to the hepatocyte host cell and also block interaction with host cell receptors on hepatocytes. In addition, specific CD4+ and CD8+ cellular mechanisms target the intracellular hepatic forms, thus preventing release of erythrocytic stage parasites from the infected hepatocyte and the ensuing blood stage cycle responsible for clinical disease. An innovative method for producing particle vaccines, layer-by-layer (LbL) fabrication of polypeptide films on solid CaCO3 cores, was used to produce synthetic malaria vaccines containing a tri-epitope CS peptide T1BT comprising the antibody epitope of the CS repeat region (B) and two T-cell epitopes, the highly conserved T1 epitope and the universal epitope T. Mice immunized with microparticles loaded with T1BT peptide developed parasite-neutralizing antibodies and malaria-specific T-cell responses including cytotoxic effector T-cells. Protection from liver stage infection following challenge with live sporozoites from infected mosquitoes correlated with neutralizing antibody levels. Although some immunized mice with low or undetectable neutralizing antibodies were also protected, depletion of T-cells prior to challenge resulted in the majority of mice remaining resistant to challenge. In addition, mice immunized with microparticles bearing only T-cell epitopes were not protected, demonstrating that cellular immunity alone was not sufficient for protective immunity. Although the microparticles without adjuvant were immunogenic and protective, a simple modification with the lipopeptide TLR2 agonist Pam3Cys increased the potency and efficacy of the LbL vaccine candidate. This study demonstrates the potential of LbL particles as promising malaria vaccine candidates using the T1BT epitopes from the P. falciparum CS protein.


Subject(s)
Antibodies, Neutralizing/immunology , Immunity, Cellular/immunology , Malaria Vaccines/chemical synthesis , Malaria Vaccines/immunology , Malaria/immunology , Malaria/prevention & control , Plasmodium falciparum/immunology , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Culicidae/parasitology , Epitopes, T-Lymphocyte/immunology , Female , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Parasites/immunology , Parasites/metabolism , Peptides/chemistry , Peptides/immunology , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Sporozoites/immunology , T-Lymphocytes/immunology , Vaccination , Vaccines, Synthetic/chemistry
7.
Vaccine ; 29(3): 558-69, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-20951665

ABSTRACT

Nanoparticle vaccines induce potent immune responses in the absence of conventional adjuvant due to the recognition by immune cells of the particle structures, which mimic natural pathogens such as viruses and bacteria. Nanoparticle vaccines were fabricated by constructing artificial biofilms using layer-by-layer (LbL) deposition of oppositely charged polypeptides and target designed peptides on CaCO(3) cores. LbL nanoparticles were efficiently internalized by dendritic cells in vitro by a mechanism that was at least partially phagocytic, and induced DC maturation without triggering secretion of inflammatory cytokines. LbL nanoparticle delivery of designed peptides to DC resulted in potent cross-presentation to CD8+ T-cells and more efficient presentation to CD4+ T-cells compared to presentation of soluble peptide. A single immunization of mice with LbL nanoparticles containing designed peptide induced vigorous T-cell responses characterized by a balanced effector (IFNγ) and Th2 (IL-4) ELISPOT profile and in vivo CTL activity. Mice immunized with LbL nanoparticles bearing ovalbumin-derived designed peptides were protected from challenge with Listeria monocytogenes ectopically expressing ovalbumin, confirming the relevance of the CTL/effector T-cell responses. LbL nanoparticles also elicited antibody responses to the target epitope but not to the matrix components of the nanoparticle, avoiding the vector or carrier affect that hampers utility of other vaccine platforms. The potency and efficacy of LbL nanoparticles administered in aqueous suspension without adjuvant or other formulation additive, and the absence of immune responses to the matrix components, suggest that this strategy may be useful in producing novel vaccines against multiple diseases.


Subject(s)
Antibodies, Bacterial/blood , Calcium Carbonate/metabolism , Nanoparticles/administration & dosage , Ovalbumin/immunology , T-Lymphocytes/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Calcium Carbonate/administration & dosage , Cross-Priming , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Female , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Ovalbumin/administration & dosage , Ovalbumin/metabolism , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL