Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Haematologica ; 108(8): 2155-2166, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36861411

ABSTRACT

Multiple Myeloma (MM) is an incurable plasma cell malignancy often treated by autologous stem cell transplant (ASCT). Clinical response to ASCT has been associated with DNA repair efficiency. Here we interrogated the role of the base excision DNA repair (BER) pathway in MM response to ASCT. Across 450 clinical samples and six disease stages, expression levels of genes in the BER pathway were found to be highly upregulated during the development of MM. In a separate cohort of 559 patients with MM treated with ASCT, expression of BER pathway members MPG and PARP3 was positively associated with overall survival (OS) while expression of PARP1, POLD1, and POLD2 was negatively associated with OS. In a validation cohort of 356 patients with MM treated with ASCT, PARP1 and POLD2 findings were replicated. In patients with MM who never received ASCT (n=319), PARP1 and POLD2 were not associated with OS, suggesting that the prognostic effect of these genes may be treatment-dependent. In preclinical models of MM, synergy was observed in anti-tumor activity when poly (ADPribose) polymerase (PARP) inhibitors (olaparib, talazoparib) were used in combination with melphalan. The negative prognosis associated with PARP1 and POLD2 expression along with the apparent melphalan-sensitizing effect of PARP inhibition may suggest this pathway as a potential biomarker in patients with MM in the setting of ASCT. Further understanding of the role of the BER pathway in MM is vital to improve therapeutic strategies related to ASCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Humans , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Melphalan/therapeutic use , Prognosis , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Transplantation, Autologous , Stem Cell Transplantation , Retrospective Studies , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/therapeutic use , DNA Polymerase III
2.
Cancer Genomics Proteomics ; 20(1): 9-17, 2023.
Article in English | MEDLINE | ID: mdl-36581339

ABSTRACT

BACKGROUND/AIM: There exists considerably large interpatient variability in pharmacokinetic exposure of high dose melphalan in multiple myeloma patients with hematopoietic stem-cell transplantation. In this study, we aimed to evaluate the potential impacts of CYP3A4*1B (rs2940574) and CYP3A5*3 (rs776746) variations on pharmacokinetic properties of melphalan and clinical outcomes in multiple myeloma (MM) patients. PATIENTS AND METHODS: Genotypes of CYP3A4*1B (rs2940574) and CYP3A5*3 (rs776746) were determined by validated gene-specific real-time PCR (RT-PCR) assays using DNA samples from 108 MM patients; plasma concentrations of melphalan at different time points were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS: CYP3A4*1B/*1B and CYP3A5*3/*3 carriers appeared to have a short median progression-free survival time and a higher maximum melphalan plasma concentration than non-carriers [792 vs. over 950 days, p=0.08; 9.91 (2.67, 34.03) vs. 8.66 (4.46, 17.61) mg/l, p=0.039]. CONCLUSION: CYP3A4*1B/*1B and CYP3A5*3/*3 variations might influence melphalan therapy in MM patients through yet-to-be-identified mechanisms.


Subject(s)
Cytochrome P-450 CYP3A , Multiple Myeloma , Humans , Cytochrome P-450 CYP3A/genetics , Chromatography, Liquid , Melphalan/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Tandem Mass Spectrometry , Genotype , Stem Cell Transplantation
3.
Front Pharmacol ; 14: 1334440, 2023.
Article in English | MEDLINE | ID: mdl-38259277

ABSTRACT

Introduction: Polymorphisms in genes responsible for the metabolism and transport of tacrolimus have been demonstrated to influence clinical outcomes for patients following allogeneic hematologic stem cell transplant (allo-HSCT). However, the clinical impact of germline polymorphisms specifically for oral formulations of tacrolimus is not fully described. Methods: To investigate the clinical impact of genetic polymorphisms in CYP3A4, CYP3A5, and ABCB1 on oral tacrolimus pharmacokinetics and clinical outcomes, we prospectively enrolled 103 adult patients receiving oral tacrolimus for the prevention of graft-versus-host disease (GVHD) following allo-HSCT. Patients were followed in the inpatient and outpatient phase of care for the first 100 days of tacrolimus therapy. Patients were genotyped for CYP3A5 *3 (rs776746), CYP3A4 *1B (rs2740574), ABCB1 exon 12 (rs1128503), ABCB1 exon 21 (rs2032582), ABCB1 exon 26 (rs1045642). Results: Expression of CYP3A5 *1 was highly correlated with tacrolimus pharmacokinetics in the inpatient phase of care (p < 0.001) and throughout the entirety of the study period (p < 0.001). Additionally, Expression of CYP3A5 *1 was associated with decreased risk of developing AKI as an inpatient (p = 0.06). Variants in ABCB1 were not associated with tacrolimus pharmacokinetics in this study. We were unable to discern an independent effect of CYP3A4 *1B or *22 in this population. Conclusion: Expression of CYP3A5 *1 is highly influential on the pharmacokinetics and clinical outcomes for patients receiving oral tacrolimus as GVHD prophylaxis following allo-HSCT.

4.
Anticancer Res ; 42(1): 385-395, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34969749

ABSTRACT

BACKGROUND: It has been reported that expression of OCT3 enhanced the sensitivity to melphalan in cells, indicative of potential roles of OCT3 in melphalan transport. Herein we investigated the association of select single nucleotide polymorphisms in SLC22A3 (gene encoding OCT3) with clinical outcomes in multiple myeloma (MM) patients with hematopoietic autologous stem cell transplants followed by high-dose melphalan therapy. MATERIALS AND METHODS: Melphlan concentrations in blood samples from 108 MM patients were measured using liquid chromatography-tandem mass spectrometry (LC-MS/ΜS); genotypes of rs2048327, rs1810126, and rs3088442 in these patients were determined using quatitive RT-PCR assays. RESULTS: Rs3088442 A variant-carriers had a significantly increased risk of severe oral mucositis in comparison with homozygous rs3088442 G-carriers with adjusted odds ratio of 4.00 (95% CI=1.25-14.7; p=0.027). Rs3088442 A carriers tended to have lower creatinine clearance (p=0.10) and higher maximum plasma concentration of melphalan (p=0.07). CONCLUSION: OCT3 might be involved in melphalan transport in MM patients.


Subject(s)
Genetic Predisposition to Disease , Multiple Myeloma/therapy , Organic Cation Transport Proteins/genetics , Stomatitis/genetics , Adult , Aged , Genetic Association Studies , Genotype , Humans , Male , Melphalan/adverse effects , Melphalan/therapeutic use , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Polymorphism, Single Nucleotide/genetics , Stem Cell Transplantation/adverse effects , Stomatitis/epidemiology , Stomatitis/pathology , Transplantation, Autologous/adverse effects
5.
Mol Carcinog ; 58(12): 2327-2339, 2019 12.
Article in English | MEDLINE | ID: mdl-31544312

ABSTRACT

Autologous stem cell transplant (ASCT) with high-dose melphalan (HDM) is the standard treatment for fit multiple myeloma (MM) patients. It is generally believed that some DNA repair proteins impact the activity to repair melphalan-induced DNA damage, thus potentially contributing to the patient's clinical response. However, knowledge of these proteins is limited. In the current study, we investigated the roles of XRCC1, a protein involved in base excision repair and single-strand break repair, in melphalan response in MM cells. Small interfering RNA knockdown of XRCC1 significantly increased the accumulation of melphalan-induced DNA damage in MM cells and sensitized them to melphalan treatment, indicating that genetic variation in XRCC1 may impact response to melphalan treatment. We then evaluated the association between an XRCC1 variant with reduced activity, rs25487 (R399Q), and clinical outcomes of 108 MM patients with melphalan therapy. Our results showed that XRCC1 rs25487 was associated with prolonged progression-free survival (PFS) in MM patients. The adjusted hazard ratio for PFS between patients carrying rs25487 AA/AG and GG was 0.42 (95% confidence interval: 0.25, 0.84, P = .014). Taken together, these results indicate that XRCC1 is involved in the repair of melphalan-induced DNA damage and XRCC1 rs25487 variant with impaired DNA repair function influences the clinical responses of HDM in MM patients.


Subject(s)
DNA Repair , Hematopoietic Stem Cell Transplantation/methods , Melphalan/therapeutic use , Multiple Myeloma/therapy , X-ray Repair Cross Complementing Protein 1/metabolism , Aged , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , DNA Breaks, Single-Stranded/drug effects , DNA Damage , Dose-Response Relationship, Drug , Female , Humans , Kaplan-Meier Estimate , Male , Melphalan/adverse effects , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Polymorphism, Single Nucleotide , Progression-Free Survival , RNA Interference , Transplantation, Autologous , X-ray Repair Cross Complementing Protein 1/genetics
6.
Bioanalysis ; 11(6): 471-483, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30892061

ABSTRACT

Aim: To develop and validate a reliable, robust and efficient assay to detect and quantify biologic compounds in vitro and in vivo during early stage of a biotherapeutic agent discovery. Methodology & results: An enrichment-free immunoassay method was developed to quantify a polyhistidine N- and FLAG C-terminally-tagged recombinant protein of ∼55 kDa. The target proteins were purified by a nickel-based matrix via tag affinity, followed by probing with biotinylated antitag antibody and subsequently detected by streptavidin-horseradish peroxidase conjugate using an automated capillary-based western system. Conclusion: A simple, highly sensitive and efficient immunoassay protocol was established to assess the in vitro stability and pharmacokinetic properties of propitious recombinant proteins in vivo in mouse to support early stage development of a biotherapeutic lead.


Subject(s)
Epitopes/chemistry , Immunoassay/methods , Recombinant Proteins/blood , Animals , Biotinylation , Blotting, Western/methods , Histidine/chemistry , Indicators and Reagents/chemistry , Male , Mice, Inbred BALB C , Mice, Inbred ICR , Nickel/chemistry , Oligopeptides/blood , Oligopeptides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Streptavidin/chemistry
7.
Anticancer Res ; 39(1): 67-72, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30591441

ABSTRACT

BACKGROUND/AIM: SLC7A5 is recognized as the major mediator of melphalan uptake into multiple myeloma (MM) cells; however, its contribution to the inter-patient variability of melphalan efficacy and toxicity is yet to be well elucidated. This study aimed to investigate the impact of a single nucleotide polymorphism (SNP) rs4240803 in SLC7A5 on the gene expression, ex vivo sensitivity to melphalan, and clinical outcomes in MM patients who were undergoing autologous stem cell transplantation with high-dose melphalan. MATERIALS AND METHODS: Peripheral blood mononuclear cells (PBMC) were collected from 108 MM patients prior to melphalan therapy. Clinical data were also collected from these patients following melphalan therapy. RESULTS: rs4240803 was associated with elevated expression of SLC7A5 mRNA, higher ex vivo sensitivity to melphalan in PBMCs, and positive 90-day response in these patients (p=0.047, 0.10, 0.049, respectively). CONCLUSION: rs4240803 impacted the expression of SLC7A5, thus contributing to the clinical response of MM patients to melphalan therapy.


Subject(s)
Large Neutral Amino Acid-Transporter 1/genetics , Melphalan/administration & dosage , Multiple Myeloma/drug therapy , Adult , Aged , Combined Modality Therapy , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukocytes, Mononuclear/drug effects , Male , Melphalan/adverse effects , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Polymorphism, Single Nucleotide , Transplantation, Autologous/adverse effects , Treatment Outcome
8.
DNA Repair (Amst) ; 57: 50-55, 2017 09.
Article in English | MEDLINE | ID: mdl-28688373

ABSTRACT

The presence of an enhancer element, RDINK4/ARF (RD), in the prominent INK4-ARF locus provides a novel en bloc mechanism to simultaneously regulate the transcription of the p15INK4B (p15), p16INK4A (p16), and p14ARF tumor suppressor genes. While genetic inactivation of p15, p16, and p14ARF in human cancers has been extensively studied, little is known about RD alteration and its potential contributions to cancer progression. In this review, we discuss recent developments in RD alteration and its association with p15, p16, and p14ARF alterations in human cancers, and demonstrate that RD deletion may represent a novel mechanism to simultaneously down-regulate p15, p16, and p14ARF, thus promoting carcinogenesis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Enhancer Elements, Genetic/genetics , Neoplasms/genetics , Sequence Deletion , Tumor Suppressor Protein p14ARF/genetics , Base Sequence , Carcinogenesis , Female , Humans , Male , Neoplasms/metabolism , Neoplasms/pathology
9.
Mol Carcinog ; 56(7): 1722-1732, 2017 07.
Article in English | MEDLINE | ID: mdl-28150872

ABSTRACT

Multiple myeloma (MM) is a hematologic malignancy characterized by clonal proliferation of plasma cells and overproduction of monoclonal immunoglobins. Treatment with melphalan is currently standard of care for younger and fit patients when followed by hematopoietic stem cell transplantation (HSCT), and in transplant ineligible patients when used in combination regimens. It has been previously shown that changes in the p53 pathway are associated with melphalan efficacy, but the regulatory role of the p14ARF-MDM2-p53 axis has yet to be fully explored. Recently, a non-coding RNA, ANRIL (antisense non-coding RNA in the INK4-ARF locus) has been shown to negatively regulate the transcription of the entire INK4-ARF locus and simultaneously modulate the p53 and pRb pathways. Moreover, some single nucleotide polymorphisms (SNPs) in ANRIL have previously been associated with susceptibility to several malignancies. Here we investigated select ANRIL SNPs in DNA from patient-derived peripheral blood mononuclear cells obtained from 108 MM patients treated with high-dose melphalan followed by HSCT. Our results show that the rs2151280 (CàT) SNP in ANRIL was associated with worse progression-free survival (TC/CC vs TT: HR = 0.53, 95%CI, [0.26, 1.07], P = 0.07; adjusted HR = 0.39, 95%CI, [0.18, 0.84], P = 0.016), and the TT variant had higher ANRIL expression and lower p15, p14ARF, and p16 expression compared to the TC/CC variants. Our results indicate that ANRIL may be involved in melphalan-mediated apoptosis via down-regulating p14ARF and subsequent p53, and that the rs2151280 polymorphism may be a potential prognostic biomarker for relapse in melphalan-treated MM patients.


Subject(s)
Leukocytes, Mononuclear/pathology , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/pathology , Polymorphism, Single Nucleotide/genetics , RNA, Long Noncoding/genetics , Stem Cell Transplantation/adverse effects , Adult , Aged , Antineoplastic Agents, Alkylating/adverse effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Combined Modality Therapy , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Follow-Up Studies , Humans , Leukocytes, Mononuclear/metabolism , Male , Melphalan/adverse effects , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Prognosis , Prospective Studies , Survival Rate , Transplantation, Autologous , Tumor Suppressor Protein p14ARF/metabolism
10.
Am J Health Syst Pharm ; 73(17): 1331-7, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27543577

ABSTRACT

PURPOSE: The stability of extemporaneously prepared erlotinib, lapatinib, and imatinib oral liquid dosage forms using two commercially available vehicles when stored at 4 and 25 °C was evaluated. METHODS: Three batches of extemporaneous oral suspensions were prepared for each drug. Erlotinib and lapatinib tablets were crushed and mixed in a 1:1 mixture of Ora-Plus:Ora-Sweet solution to yield 10- and 50-mg/mL suspensions, respectively. Imatinib tablets were crushed and mixed in Ora-Sweet solution to yield a 40-mg/mL suspension. Suspensions were stored in amber plastic bottles, and samples from each bottle were obtained on days 0, 1, 3, 7, 14, and 28. RESULTS: Erlotinib 10-mg/mL and lapatinib 50-mg/mL oral suspensions in a 1:1 mixture of Ora-Plus and Ora-Sweet retained at least 90% of their initial concentration throughout the 28-day study when stored at 25 °C. Visual inspection revealed notable viscosity changes in the erlotinib and lapatinib suspensions stored at 4 °C for 7 days and beyond. The viscosity of these preparations increased with time and was particularly evident with the erlotinib suspension, which exhibited a puddinglike texture. Imatinib 40-mg/mL oral suspension in Ora-Sweet appeared stable for up to 14 days when stored at both 25 and 4 °C. CONCLUSION: Erlotinib 10-mg/mL and lapatinib 50-mg/mL oral suspensions prepared from commercially available tablets were stable for at least 28 days when prepared in a 1:1 mixture of Ora-Plus:Ora-Sweet at 25 °C. Imatinib 40-mg/mL oral suspension prepared from commercially available tablets was stable for up to 14 days when prepared in Ora-Sweet and stored at 25 and 4 °C.


Subject(s)
Chemistry, Pharmaceutical/methods , Erlotinib Hydrochloride/chemistry , Imatinib Mesylate/chemistry , Quinazolines/chemistry , Administration, Oral , Chemistry, Pharmaceutical/standards , Dosage Forms , Drug Stability , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/standards , Humans , Imatinib Mesylate/administration & dosage , Imatinib Mesylate/standards , Lapatinib , Quinazolines/administration & dosage , Quinazolines/standards , Suspensions
11.
Mol Carcinog ; 54(7): 532-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24302590

ABSTRACT

The presence of RD(INK4/ARF) (RD) enhancer in the INK4-ARF locus provides a novel mechanism to simultaneously increase the transcription of p15(INK4b) (p15), p14ARF (p14), and p16(INK4a) (p16). While such upregulation can be repressed through interactions between RD and oncoproteins CDC6 and BMI1, little is known about the involvement of RD in cancer. In this study we investigated RD deletions in 30 squamous cell carcinoma of the head and neck (SCCHN) and the patient-matched High At-Risk Mucosa specimens (HARM, "phenotypically normal" tissues neighboring SCCHN foci but beyond the surgical resection margin). RD was deleted (homozygously/heterozygously) in SCCHN and HARM at the incidence of 36.7% (11/30) and 13.3% (4/30), respectively. In comparison, no RD deletion was detected in 26 oral buccal brush biopsy specimens from healthy donors. Both p16 and p14 were lowly expressed in SCCHN and HARM, and their mRNA expression levels were positively associated with each other (P < 0.01). Moreover, BMI1 was highly expressed in both SCCHN and HARM, and BMI1 overexpression was associated with p16 downregulation in SCCHN (P < 0.05). These results indicate that RD deletion and BMI1 overexpression frequently occur in the early stage of oral carcinogenesis and BMI1 overexpression may downregulate the transcription of p16 and p14 through interfering with RD.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Regulation, Neoplastic , Genetic Loci , Head and Neck Neoplasms/genetics , Tumor Suppressor Protein p14ARF/genetics , Female , Gene Deletion , Humans , Male , Middle Aged , Polycomb Repressive Complex 1/genetics , Squamous Cell Carcinoma of Head and Neck
12.
Anticancer Res ; 34(7): 3285-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24982332

ABSTRACT

BACKGROUND/AIM: While aberrant expression of cyclin-dependent kinase-4 (CDK4) has been found in squamous cell carcinoma of the head and neck (SCCHN), the associations between CDK4 and its regulators, namely, cyclin D1, cyclin E, gankyrin, SEI1, and BMI1 in gene expression remain to be explored. Herein we investigated the mRNA profiles of these oncogenes and their interrelations in different oral lesion tissues. MATERIALS AND METHODS: Thirty SCCHN specimens and patient-matched high at-risk mucosa (HARM) and 16 healthy control specimens were subjected to quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analyses. RESULTS: The mRNA levels of CDK4, cyclin D1, gankyrin, SEI1, BMI1 were significantly elevated in both HARM and SCCHN (in comparison with control specimens), and statistically significant correlations were found among these markers in gene expression. CONCLUSION: Up-regulation of CDK4 and its regulators takes place in oral cancer progression in a coordinate manner, and HARM and SCCHN share a similar molecular signature within the CDK4-pRB pathway.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Cyclin-Dependent Kinase 4/biosynthesis , Head and Neck Neoplasms/enzymology , Mouth Neoplasms/enzymology , Adult , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin E/biosynthesis , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinase 7/biosynthesis , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Principal Component Analysis , Proteasome Endopeptidase Complex/biosynthesis , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Squamous Cell Carcinoma of Head and Neck , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Up-Regulation
13.
Pancreas ; 43(7): 1009-13, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25003221

ABSTRACT

OBJECTIVE: The presence of an enhancer element, RD (RD), in the prominent INK4-ARF locus provides a novel en bloc mechanism to simultaneously regulate the transcription of p15, p14ARF, and p16 genes. However, knowledge about RD alterations and its potential contributions to cancer progression remains limited. In this study, we aimed to evaluate the incidence of RD alterations in pancreatic tumors. METHODS: DNAs from 14 gastrinomas and 6 nonfunctioning pancreatic neuroendocrine tumors were subjected to quantitative real-time polymerase chain reaction-based assays to determine deletions in p15, p14ARF, and p16 (both exons 1 and 2). RESULTS: RD was frequently deleted in gastrinomas and nonfunctioning pancreatic neuroendocrine tumors with an incidence of 30% (6/20 samples). In comparison, the incidences of deletions of p15 (exon 1), p14ARF (exon 1ß), and p16 (exon 1α) are 10% (2/20 samples), 10% (2/20 samples), and 45% (9/20 samples), respectively. Whereas some RD deletion events arose from deletions of the entire INK4-ARF locus, RD deletions in some specimens seemed to be independent of genetic alterations in any of the p15, p14ARF, and p16 genes. CONCLUSIONS: Our results strongly support that the deletion of RD may represent a novel mechanism to simultaneously downregulate p15, p14ARF, and p16, thus contributing to the development of human pancreatic cancers.


Subject(s)
Enhancer Elements, Genetic/genetics , Gastrinoma/genetics , Neoplasm Proteins/genetics , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Sequence Deletion , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Exons/genetics , Gastrinoma/physiopathology , Gene Deletion , Genes, p16/physiology , Humans , Neuroendocrine Tumors/physiopathology , Pancreatic Neoplasms/physiopathology , Real-Time Polymerase Chain Reaction , Tumor Suppressor Protein p14ARF/genetics
14.
Hum Mol Genet ; 23(1): 268-78, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23985325

ABSTRACT

Cytochrome P450 2D6 (CYP2D6) is involved in the metabolism of 25% of clinically used drugs. Genetic polymorphisms cause substantial variation in CYP2D6 activity and serve as biomarkers guiding drug therapy. However, genotype-phenotype relationships remain ambiguous except for poor metabolizers carrying null alleles, suggesting the presence of yet unknown genetic variants. Searching for regulatory CYP2D6 polymorphisms, we find that a SNP defining the CYP2D6*2 allele, rs16947 [R296C, 17-60% minor allele frequency (MAF)], previously thought to convey normal activity, alters exon 6 splicing, thereby reducing CYP2D6 expression at least 2-fold. In addition, two completely linked SNPs (rs5758550/rs133333, MAF 13-42%) increase CYP2D6 transcription more than 2-fold, located in a distant downstream enhancer region (>100 kb) that interacts with the CYP2D6 promoter. In high linkage disequilibrium (LD) with each other, rs16947 and the enhancer SNPs form haplotypes that affect CYP2D6 enzyme activity in vivo. In a pediatric cohort of 164 individuals, rs16947 alone (minor haplotype frequency 28%) was associated with reduced CYP2D6 metabolic activity (measured as dextromethorphan/metabolite ratios), whereas rs5758550/rs133333 alone (frequency 3%) resulted in increased CYP2D6 activity, while haplotypes containing both rs16947 and rs5758550/rs133333 were similar to the wild-type. Other alleles used in biomarker panels carrying these variants such as CYP2D6*41 require re-evaluation of independent effects on CYP2D6 activity. The occurrence of two regulatory variants of high frequency and in high LD, residing on a long haplotype, highlights the importance of gene architecture, likely shaped by evolutionary selection pressures, in determining activity of encoded proteins.


Subject(s)
Alternative Splicing , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Promoter Regions, Genetic , Transcription, Genetic , Exons , Genetic Loci , Genetic Variation , Genotype , Haplotypes , Hep G2 Cells , Humans , Linkage Disequilibrium , Liver/metabolism , Phenotype , Polymorphism, Single Nucleotide
15.
Mol Carcinog ; 53(3): 211-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23065809

ABSTRACT

Recent identification of an enhancer element, RD(INK4/ARF) (RD), in the prominent INK4/ARF locus provides a novel mechanism to simultaneously regulate the transcription of p15(INK4B) (p15), p14(ARF) , and p16(INK4A) (p16) tumor suppressor genes. While genetic inactivation of p15, p14(ARF) , and p16 in human tumors has been extensively studied, little is known about genetic alterations of RD and its impact on p15, p14(ARF) , and p16 in human cancer. The purpose of this study was to investigate the potential existence of genetic alterations of RD in human cancer cells. DNAs extracted from 17 different cancer cell lines and 31 primary pheochromocytoma tumors were analyzed for deletion and mutation of RD using real-time PCR and direct DNA sequencing. We found that RD was deleted in human cancer cell lines and pheochromocytoma tumors at frequencies of 41.2% (7/17) and 13.0% (4/31), respectively. While some of these RD deletion events occurred along with deletions of the entire INK4/ARF locus, other RD deletion events were independent of genetic alterations in p15, p14(ARF) , and p16. Furthermore, the status of RD was poorly associated with the expression of p15, p14(ARF) , and p16 in tested cancer cell lines and tumors. This study demonstrates for the first time that deletion of the RD enhancer is a prevalent event in human cancer cells. Its implication in carcinogenesis remains to be further explored.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Enhancer Elements, Genetic/genetics , Neoplasms/genetics , Point Mutation/genetics , Tumor Suppressor Protein p14ARF/genetics , Adrenal Gland Neoplasms/genetics , DNA Methylation , Gene Deletion , Humans , Pheochromocytoma/genetics , Tumor Cells, Cultured
16.
Biochem Biophys Res Commun ; 436(2): 217-22, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23727582

ABSTRACT

The INK4a-ARF locus plays a central role in the development of pancreatic tumors as evidenced by the fact that up to 98% of pancreatic tumor specimens harbored genetic alterations at the INK4a-ARF locus. Interestingly, in addition to the well-known P16(INK4A) (P16) and P14ARF tumor suppressors, the INK4a-ARF locus in pancreas encodes another protein, P12, whose structure, function, and contributions to pancreatic carcinogenesis remain to be elucidated. In the current study, we demonstrated that over-expression of p12 in human pancreatic cancer cells led to cell arrest at the G1 phase and such cell cycle arrest was related to down-regulation of a number of oncogenes, such as c-Jun, Fos, and SEI1. Furthermore, unlike P16, P12 did not retain any cyclin-dependent kinase 4 (CDK4)-inhibitory activity. Instead, P12 exhibited a transactivating activity not found in P16. We also examined the genetic status of p12 in a cohort of 40 pancreatic tumor specimens and found that p12 alteration was prevalent in pancreatic tumors with an incidence of 70% (28/40). These results support that P12 is a tumor suppressive protein distinct from P16, and its genetic inactivation is associated with pancreatic carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Pancreatic Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Alternative Splicing , Amino Acid Sequence , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Transcriptional Activation , Tumor Suppressor Proteins/metabolism
17.
Support Care Cancer ; 21(10): 2679-86, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23686402

ABSTRACT

PURPOSE: As the result of a recent national shortage in paclitaxel, some patients who were receiving or scheduled to receive weekly paclitaxel were converted to every 3-week (q3w) docetaxel with granulocyte colony-stimulating factor support. Our institution noted higher than expected incidence of severe skin toxicity events attributable to docetaxel during the shortage period among our breast cancer patients. In this report, we summarize the clinical course of the first five cases, review the literature surrounding docetaxel-induced skin toxicity, and offer possible prevention and treatment strategies to improve docetaxel tolerability. METHODS: The observation period for this case series was August 1 through October 21, 2011. All patients treated with docetaxel were identified from our electronic medical record. Operable stage I-III breast cancer patients who received ≥ 1 dose of docetaxel monotherapy at 75-100 mg/m(2) q3w were included in this study. The cases of grade 3-4 docetaxel-induced skin toxicities identified by the treating oncologists were then contacted and signed an informed consent through an Institutional Review Board-approved protocol. RESULTS: Thirty-four patients met the inclusion criteria. Five patients (14.7 %) experienced grade 3 skin toxicity events attributable to docetaxel, a significantly higher rate than previously reported for docetaxel dosed at 75-100 mg/m(2). CONCLUSIONS: Docetaxel-induced dermatologic toxicity is well characterized; nonetheless, its etiology is largely unknown and evidence-based prevention and management strategies are lacking. This report shows that the use of docetaxel 75-100 mg/m(2) q3w subsequent to dose-dense doxorubicin and cyclophosphamide regimen can lead to unacceptable rate of severe skin toxicity.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/supply & distribution , Breast Neoplasms/drug therapy , Paclitaxel/supply & distribution , Skin Diseases/chemically induced , Taxoids/adverse effects , Antineoplastic Agents/administration & dosage , Docetaxel , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Incidence , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Survival Rate , Taxoids/administration & dosage
18.
Clin Pharmacokinet ; 52(8): 705-12, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23588536

ABSTRACT

BACKGROUND AND OBJECTIVE: Pentostatin is an irreversible inhibitor of adenosine deaminase and has been used to prevent graft-versus-host disease (GVHD) and to treat both acute and chronic GVHD. Dose reduction equations for patients with renal insufficiency are based on few patients with limited pharmacokinetic and clinical results. This phase II study (NCT00201786) was conducted to assess pentostatin efficacy and infectious complications seen from our previous phase I study in steroid-refractory acute GVHD (aGVHD). PATIENTS AND METHODS: Hospitalized patients with steroid-refractory aGVHD were given pentostatin 1.5 mg/m(2)/day intravenously on days 1-3 of each 14-day cycle. Prior to each dose, dose modifications were based on Cockcroft-Gault estimated creatinine clearance (eCrCL) with 30-50 mL/min/1.73 m(2) leading to a 50 % dose reduction and eCrCL less than 30 mL/min/1.73 m(2) leading to study removal. Plasma pentostatin area under the concentration-time curve (AUC) and incidence of infectious complications were evaluated. RESULTS: Two of the eight patients treated demonstrated excessive pentostatin exposure as determined by measurement of AUC. One of these patients had renal impairment, whereas the other patient demonstrated borderline renal function. Despite dose reduction to 0.75 mg/m(2), AUCs were significantly increased compared to the other patients in this study. Seven of eight patients treated with pentostatin had cytomegalovirus (CMV) viremia after pentostatin treatment; however none developed proven CMV disease. CONCLUSION: A 50 % dose reduction in patients with eCrCL 30-50 mL/min/1.73 m(2) seems reasonable. However, the eCrCL should be interpreted with extreme caution in patients who are critically ill and/or with poor performance status. Renal function assessment based on the Cockcroft-Gault method could be significantly overestimated thus risking pentostatin overdosing. These results imply a need to closely monitor pentostatin exposure in patients with renal insufficiency.


Subject(s)
Adenosine Deaminase Inhibitors/administration & dosage , Adenosine Deaminase Inhibitors/pharmacokinetics , Graft vs Host Disease/blood , Pentostatin/administration & dosage , Pentostatin/pharmacokinetics , Adenosine Deaminase Inhibitors/blood , Adult , Antibodies, Monoclonal/administration & dosage , Area Under Curve , Blood Transfusion, Autologous , Creatinine/blood , Cyclosporine/administration & dosage , Drug Resistance , Female , Graft vs Host Disease/drug therapy , Humans , Immunosuppressive Agents/administration & dosage , Infliximab , Lymphocyte Transfusion , Male , Methotrexate/administration & dosage , Methylprednisolone/therapeutic use , Middle Aged , Pentostatin/blood , Renal Insufficiency/blood , Renal Insufficiency/drug therapy , Stem Cell Transplantation , Tacrolimus/administration & dosage , Young Adult
19.
Carcinogenesis ; 25(2): 263-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14604895

ABSTRACT

The p16INK4A/CDKN2A (p16) tumor suppressor gene is known to be inactivated in up to 98% of human pancreatic cancer specimens. Chemically induced pancreatic tumors in Syrian golden hamsters have been demonstrated to share many morphological and biological similarities with human pancreatic tumors and represent a potentially suitable model for the evaluation of therapies targeting p16. The purpose of this study was to evaluate primary hamster pancreatic tumor specimens for potentially inactivating p16 alterations. Tumors were induced with N-nitroso-bis-(2-oxopropyl) amine, followed by two cycles of augmentation pressure, and were harvested on day 100. Foci of tumor cells were identified by light microscopy after staining with hematoxylin and eosin, and corresponding tumor tissues were excised for DNA extraction. The techniques of multiplex real-time PCR, direct sequencing and methylation-specific PCR were used to evaluate 30 tumor specimens for homozygous deletions, mutations and aberrant methylation of 5' CpG islands, respectively. Homozygous deletions were identified in 11 of 30 (36.7%) specimens, mutations were identified in four of 30 (13.3%) specimens, and aberrant methylation of 5' CpG islands was found in 14 of 30 (46.7%) specimens. The overall frequency of p16 alterations was 93.3% (28 of 30 specimens) and the majority of changes (83.3%) were noted to be secondary to methylation or homozygous deletion. The four mutations significantly impaired cyclin-dependent kinase 4 inhibitory activity, and two resulted in perturbation of the global structure of P16 protein. These findings indicate that p16 inactivation is a common event in chemically induced hamster tumors, and that this animal model is appropriate for comparative studies evaluating pancreatic cancer therapeutic strategies targeting p16.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , Nitrosamines/toxicity , Pancreatic Neoplasms/metabolism , Animals , CpG Islands , Cricetinae , Female , Gene Deletion , Homozygote , Mesocricetus , Mutation , Pancreatic Neoplasms/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...