Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Clin Lymphoma Myeloma Leuk ; 22(7): e526-e531, 2022 07.
Article in English | MEDLINE | ID: mdl-35361554

ABSTRACT

Selinexor is a first in class selective inhibitor of nuclear export (SINE), blocks exportin 1 (XPO1), a protein transporter, that among other actions, shuttles cargo proteins such as tumor suppressor proteins (TSPs), the glucocorticoid receptor (GR), and oncoprotein messenger RNAs (mRNAs) across the nuclear membrane to cytoplasm. By blocking XPO1, selinexor facilitates nuclear preservation and activation of TSPs, and prevents mRNA translation of the oncoproteins leading to induction of apoptosis. The therapeutic value of selinexor in combination with dexamethasone has been successfully demonstrated in treating relapsed and/or refractory myeloma (RRMM), leading to the Food and Drug Administration (FDA) approval of selinexor in combination with dexamethasone in 2019 for the treatment of adult patients with RRMM who received at least 4 prior therapies and whose disease is refractory to at least 2 proteasome inhibitors, at least 2 immunomodulatory agents, and an anti-CD38 monoclonal antibody (mAb) - a pentarefractory myeloma. More recently, selinexor in combination with bortezomib and dexamethasone was approved by the FDA in December 2020, based on the BOSTON study among RRMM patients who had received at least one prior line of therapy. With more available safety and efficacy data supporting the increased interval between dosing of selinexor (and lesser cumulative weekly dosing) and schedule, contrary to the originally approved dose of 160 mg per week, the supportive care guidelines needed to be revisited. The current manuscript summarizes the supportive care solutions with weekly dosing of selinexor and identifies the ideal potential patient for selinexor treatment.


Subject(s)
Multiple Myeloma , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Consensus , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Humans , Hydrazines/pharmacology , Hydrazines/therapeutic use , Multiple Myeloma/pathology , Triazoles
2.
Leukemia ; 32(10): 2305, 2018 10.
Article in English | MEDLINE | ID: mdl-30218008

ABSTRACT

Following the publication of this article, the authors noted that the pomalidomide dose for the additional SC cohort in Fig. 1 was incorrectly listed. The correct dose for pomalidomide in the additional SC cohort should be the maximum tolerated dose of 4 mg/day, not 2 mg/day as listed in the original Fig. 1. The authors apologize for any inconvenience caused.

3.
Leukemia ; 31(12): 2695-2701, 2017 12.
Article in English | MEDLINE | ID: mdl-28642620

ABSTRACT

This phase 1 dose-escalation study evaluated pomalidomide, bortezomib (subcutaneous (SC) or intravenous (IV)) and low-dose dexamethasone (LoDEX) in lenalidomide-refractory and proteasome inhibitor-exposed relapsed or relapsed and refractory multiple myeloma (RRMM). In 21-day cycles, patients received pomalidomide (1-4 mg days 1-14), bortezomib (1-1.3 mg/m2 days 1, 4, 8 and 11 for cycles 1-8; days 1 and 8 for cycle ⩾9) and LoDEX. Primary endpoint was to determine the maximum tolerated dose (MTD). Thirty-four patients enrolled: 12 during escalation, 10 in the MTD IV bortezomib cohort and 12 in the MTD SC bortezomib cohort. Patients received a median of 2 prior lines of therapy; 97% bortezomib exposed. With no dose-limiting toxicities, MTD was defined as the maximum planned dose: pomalidomide 4 mg, bortezomib 1.3 mg/m2 and LoDEX. All patients discontinued treatment by data cutoff (2 April 2015). The most common grade 3/4 treatment-emergent adverse events were neutropenia (44%) and thrombocytopenia (26%), which occurred more frequently with IV than SC bortezomib. No grade 3/4 peripheral neuropathy or deep vein thrombosis was reported. Overall response rate was 65%. Median duration of response was 7.4 months. Pomalidomide, bortezomib and LoDEX was well tolerated and effective in lenalidomide-refractory and bortezomib-exposed patients with RRMM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Bortezomib/administration & dosage , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Lenalidomide , Male , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Neoplasm Staging , Proteasome Inhibitors/administration & dosage , Proteasome Inhibitors/therapeutic use , Retreatment , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Treatment Outcome
4.
Leukemia ; 31(12): 2630-2641, 2017 12.
Article in English | MEDLINE | ID: mdl-28439109

ABSTRACT

Autologous stem cell transplantation (ASCT) is a standard treatment for eligible multiple myeloma (MM) patients, but many patients will relapse after ASCT and require subsequent therapy. The proteasome inhibitor carfilzomib is approved for relapsed or refractory MM (RRMM). In phase 3 trials, carfilzomib-based regimens (ASPIRE, carfilzomib-lenalidomide-dexamethasone; ENDEAVOR, carfilzomib-dexamethasone) demonstrated superior progression-free survival (PFS) compared with standard therapies for RRMM (ASPIRE: lenalidomide-dexamethasone; ENDEAVOR, bortezomib-dexamethasone). This subgroup analysis of ASPIRE and ENDEAVOR evaluated outcomes according to prior ASCT status. In total, 446 patients in ASPIRE and 538 in ENDEAVOR had prior ASCT. Median PFS was longer for carfilzomib-based regimens vs non-carfilzomib-based regimens for patients with prior ASCT (ASPIRE: 26.3 vs 17.8 months (hazard ratio (HR)=0.68); ENDEAVOR: not estimable vs 10.2 months (HR=0.61)), those with one prior line of therapy that included ASCT (ASPIRE: 29.7 vs 17.8 months (HR=0.70); ENDEAVOR: not estimable vs 11.2 months (HR=0.46)), and those without prior ASCT (ASPIRE: 26.4 vs 16.6 months (HR=0.76); ENDEAVOR: 17.7 vs 8.5 months (HR=0.43)). Overall response rates also favored the carfilzomib-based regimens. No new safety signals were detected. This analysis suggests that carfilzomib-based treatment may lead to improvement in PFS and response rates regardless of prior transplant status. Further evaluation is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Female , Hematopoietic Stem Cell Transplantation , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/therapy , Oligopeptides/administration & dosage , Postoperative Care , Recurrence , Retreatment , Transplantation, Autologous , Treatment Outcome
5.
Blood Cancer J ; 6(9): e466, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27588519

ABSTRACT

In Eastern Cooperative Oncology Group-ACRIN E4A03, on completion of four cycles of therapy, newly diagnosed multiple myeloma patients had the option of proceeding to autologous peripheral blood stem cell transplant (ASCT) or continuing on their assigned therapy lenalidomide plus low-dose dexamethasone (Ld) or lenalidomide plus high-dose dexamethasone (LD). This landmark analysis compared the outcome of 431 patients surviving their first four cycles of therapy pursuing early ASCT to those continuing on their assigned therapy. Survival distributions were estimated using the Kaplan-Meier method and compared with log-rank test. Ninety patients (21%) opted for early ASCT. The 1-, 2-, 3-, 4- and 5-year survival probability estimates were higher for early ASCT versus no early ASCT at 99, 93, 91, 85 and 80% versus 94, 84, 75, 65 and 57%, respectively. The median overall survival (OS) in the early versus no early ASCT group was not reached (NR) versus 5.78 years. In patients <65 years of age, median OS in the early versus no early ASCT groups was NR in both, hazard ratio 0.79, 95% confidence interval: (0.50, 0.25). In patients ⩾65 years of age, median OS in the early versus no early ASCT was NR versus 5.11 years. ASCT dropped out of statistical significance (P=0.080). Patients opting for ASCT after induction Ld/LD had a higher survival probability and improvement in OS regardless of dexamethasone dose density.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Multiple Myeloma/therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Clinical Trials, Phase III as Topic , Combined Modality Therapy , Dexamethasone/administration & dosage , Follow-Up Studies , Humans , Lenalidomide , Middle Aged , Multiple Myeloma/diagnosis , Multiple Myeloma/mortality , Neoplasm Staging , Proportional Hazards Models , Randomized Controlled Trials as Topic , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Transplantation, Autologous , Treatment Outcome
7.
Blood Cancer J ; 4: e182, 2014 02 21.
Article in English | MEDLINE | ID: mdl-24562384

ABSTRACT

The addition of vorinostat to lenalidomide/dexamethasone represents a novel combination therapy in multiple myeloma (MM), informed by laboratory studies suggesting synergy. This was a phase I, multicenter, open-label, non-randomized, dose-escalating study in patients with relapsed or relapsed and refractory MM. Clinical evaluation, electrocardiogram, laboratory studies and adverse events were obtained and assessed. The maximum-tolerated dose was not reached owing to a non-occurrence of two dose-limiting toxicities per six patients tested at any of the dosing levels. Patients tolerated the highest dose tested (Level 5) and this was considered the maximum administered dose: at 400 mg vorinostat on days 1-7 and 15-21, 25 mg lenalidomide on days 1-21 and 40 mg dexamethasone on days 1, 8, 15 and 22, per 28-day cycle. Drug-related adverse events were reported in 90% of patients serious adverse experiences were reported in 45% of the patients and 22% of all patients had adverse experiences considered, possibly related to study drug by the investigators. A confirmed partial response or better was reported for 14/30 patients (47%) evaluable for efficacy, including 31% of patients previously treated with lenalidomide. Vorinostat in combination with lenalidomide and dexamethasone proved tolerable with appropriate supportive care, with encouraging activity observed.

8.
Leukemia ; 27(12): 2351-6, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23670297

ABSTRACT

Several cytogenetic abnormalities are associated with poor outcomes in multiple myeloma (MM). We prospectively analyzed the impact of cytogenetic abnormalities on outcomes during the phase 2 PX-171-003-A1 study of single-agent carfilzomib for relapsed and refractory MM. In the response-evaluable population (257/266), fluorescence in situ hybridization (FISH)/conventional cytogenetic profiles were available for 229 patients; 62 (27.1%) had high-risk cytogenetics--del 17p13, t(4;14) or t(14;16) by interphase FISH or deletion 13 or hypodiploidy by metaphase cytogenetics--and 167 (72.9%) had standard-risk profiles. Generally, baseline characteristics were similar between the subgroups, but International Staging System stage III disease was more common in high- vs standard-risk patients (41.9% vs 27.5%) as was Eastern Cooperative Oncology Group performance status 1/2 (85.5% vs 68.3%). Overall response was comparable between the subgroups (25.8% vs 24.6%, respectively; P=0.85), while time-to-event end points showed a trend of shorter duration in high-risk patients, including median duration of response (5.6 months (95% confidence interval (CI) 3.7-7.8) vs 8.3 months (95% CI 5.6-12.3)) and overall survival (9.3 (95% CI 6.5-13.0) vs 19.0 months (95% CI 15.4-NE); P=0.0003). Taken together, these findings demonstrate that single-agent carfilzomib is efficacious and has the potential to at least partially overcome the impact of high-risk cytogenetics in heavily pre-treated patients with MM.


Subject(s)
Antineoplastic Agents/therapeutic use , Chromosome Aberrations , Multiple Myeloma/drug therapy , Oligopeptides/therapeutic use , Adult , Aged , Aged, 80 and over , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Multiple Myeloma/genetics , Survival Analysis , Treatment Outcome
10.
Leuk Lymphoma ; 46(8): 1137-42, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16085553

ABSTRACT

Malignant pleural effusion (MPE) in multiple myeloma (MM) is rare. Approximately 80 cases have been reported. To delineate optimal treatment and prognostic variables in these patients, we reviewed 11 MM patients with MPE. MPE developed at median of 12 months from diagnosis of MM. All the patients had high-risk disease based on complex karyotypic abnormalities including deletions of chromosome-13 (n=9), elevated beta-2 microglobulin (B2M) (n=9), high C-reactive protein (CRP) (n=8), high plasma cell labeling index (n=5) or high LDH (n=5). A significant increase in B2M, LDH, and CRP was observed at the onset of MPE. The initial diagnosis of MPE was based on positive cytology (n=9), pleural fluid cIg/DNA (n=9) or pleural fluid cytogenetics (n=4). Pleural tissue infiltration was found on pleural biopsy and autopsy in one patient each. Systemic chemotherapy comprising dexamethasone, cyclophosphamide, etoposide and cisplatin (DCEP) (n=7) and pleurodesis (n=7) were effective in resolving MPE but survival was short. High dose chemotherapy with peripheral blood stem cell support for MPE in six patients conferred no clear survival advantage. These patients died at median of four months from onset of MPE. Patients with bone marrow complex karyotypic abnormalities including deletion-13 (n=9) had a shorter (median--18 months) overall survival compared to patients with normal cytogenetics (median--38 months). MPE in patients with MM is often associated with high-risk disease including deletion 13 chromosomal abnormality and heralds a poor prognosis despite aggressive local and systemic treatment.


Subject(s)
C-Reactive Protein/metabolism , L-Lactate Dehydrogenase/metabolism , Multiple Myeloma/complications , Multiple Myeloma/diagnosis , Pleural Effusion, Malignant/complications , Pleural Effusion, Malignant/diagnosis , beta 2-Microglobulin/metabolism , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Karyotyping , Male , Middle Aged , Multiple Myeloma/drug therapy , Pleural Effusion, Malignant/drug therapy , Predictive Value of Tests , Prognosis , Survival Analysis , Treatment Outcome
11.
Int J Occup Environ Health ; 5(2): 127-35, 1999.
Article in English | MEDLINE | ID: mdl-10330514

ABSTRACT

The efforts of workers in less-developed countries who have been exposed to 1,2-dibromo-3-chloropropane (DBCP) to obtain redress through the courts for damages suffered from these exposures are reported. The authors, who are lawyers, have represented more than 26,000 such workers. Evidence of the culpability of the U.S. manufacturers and the corporate users of DBCP, particularly Standard Fruit Company in Costa Rica, is presented. The damaged-worker plaintiffs are stymied by the application by the U.S. judicial system of forum non conveniens, which works in the defendants' favor by shunting the cases back to the plaintiffs' home countries, where the judicial systems are inadequate to deal with such cases and unlikely to be able to enforce judgments against the defendants.


Subject(s)
Developing Countries , Industry/legislation & jurisprudence , Insecticides/history , Occupational Exposure/legislation & jurisprudence , Propane/analogs & derivatives , History, 20th Century , Humans , Insecticides/adverse effects , Insecticides/economics , International Cooperation , Liability, Legal , Propane/adverse effects , Propane/economics , Propane/history , United States
12.
Blood ; 93(9): 3044-52, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10216101

ABSTRACT

The molecular mechanisms by which multiple myeloma (MM) cells evade glucocorticoid-induced apoptosis have not been delineated. Using a human IgAkappa MM cell line (ARP-1), we found that dexamethasone (Dex)-induced apoptosis is associated with decreased NF-kappaB DNA binding and kappaB-dependent transcription. Both nuclear p50:p50 and p50:p65 NF-kappaB complexes are detected in ARP-1 cells by supershift electrophoretic mobility shift assay (EMSA). Dex-mediated inhibition of NF-kappaB DNA binding precedes a notable increase in annexin V binding, thereby indicating that diminished NF-kappaB activity is an early event in Dex-induced apoptosis. Overexpression of bcl-2 in ARP-1 cells prevents Dex-mediated repression of NF-kappaB activity and apoptosis. Sustained NF-kappaB DNA binding is also observed in two previously characterized Dex-resistant MM cell lines (RPMI8226 and ARH-77) that express moderate levels of endogenous bcl-2 and IkappaBalpha proteins. In addition, enforced bcl-2 expression in ARP-1 cells did not prevent the augmentation of IkappaBalpha protein by Dex. We also noted a possible association between Dex-mediated downregulation of NF-kappaB in freshly obtained primary myeloma cells and the patients' responsiveness to glucocorticoid-based chemotherapy. Collectively, our data suggest that the protective effects of bcl-2 in MM cells act upstream in the NF-kappaB activation-signaling pathway and the potential use of NF-kappaB as a biomarker in progressive MM.


Subject(s)
Apoptosis/physiology , Dexamethasone/pharmacology , Genes, bcl-2 , Glucocorticoids/pharmacology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Apoptosis/drug effects , Bone Marrow/pathology , DNA, Neoplasm/metabolism , Humans , Luciferases/genetics , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured
13.
Blood ; 93(1): 51-4, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9864145

ABSTRACT

Multiple myeloma (MM) typically afflicts elderly patients with a median age of 65 years. However, while recently shown to provide superior outcome to standard treatment, high-dose therapy (HDT) has usually been limited to patients up to 65 years. Among 550 patients with MM and a minimum follow-up of 18 months, 49 aged >/=65 years were identified (median age, 67; range, 65 to 76 years). Their outcome was compared with 49 younger pair mates (median, 52; range, 37 to 64 years) selected among the remaining 501 younger patients (<65 years) matched for five previously recognized critical prognostic factors (cytogenetics, beta2-microglobulin, C-reactive protein, albumin, creatinine). Nearly one half had been treated for more than 1 year with standard therapy and about one third had refractory MM. All patients received high-dose melphalan-based therapy; 76% of the younger and 65% of the older group completed a second transplant (P =.3). Sufficient peripheral blood stem cells to support two HDT cycles (CD34 > 5 x 10(6)/kg) were available in 83% of younger and 73% of older patients (P =.2). After HDT, hematopoietic recovery to critical levels of granulocytes (>500/microL) and of platelets (>50,000/microL) proceeded at comparable rates among younger and older subjects with both first and second HDT. The frequency of extramedullary toxicities was comparable. Treatment-related mortality with the first HDT cycle was 2% in younger and 8% among older subjects, whereas no mortality was encountered with the second transplant procedure. Comparing younger/older subjects, median durations of event-free and overall survival were 2.8/1.5 years (P =.2) and 4.8/3.3 years (P =.4). Multivariate analysis showed pretransplant cytogenetics and beta2-microglobulin levels as critical prognostic features for both event-free and overall survival, whereas age was insignificant for both endpoints (P =.2/.8). Thus, age is not a biologically adverse parameter for patients with MM receiving high-dose melphalan-based therapy with peripheral blood stem cell support and, hence, should not constitute an exclusion criterion for participation in what appears to be superior therapy for symptomatic MM.


Subject(s)
Aging/physiology , Bone Marrow Transplantation/statistics & numerical data , Multiple Myeloma/physiopathology , Multiple Myeloma/therapy , Adult , Aged , Blood Platelets/drug effects , Blood Platelets/radiation effects , Combined Modality Therapy/statistics & numerical data , Granulocytes/drug effects , Granulocytes/radiation effects , Humans , Middle Aged , Multiple Myeloma/blood , Prognosis , Survival Rate , Transplantation, Autologous/statistics & numerical data
14.
Proc Natl Acad Sci U S A ; 95(1): 162-6, 1998 Jan 06.
Article in English | MEDLINE | ID: mdl-9419346

ABSTRACT

Multiple myeloma (MM) is a B cell malignancy characterized by the expansion of monoclonal Ig-secreting plasma cells with low proliferative activity. It is postulated that inhibition of physiologic cell death is an underlying factor in the pathophysiology of MM. The development of chemoresistance is a common feature in patients with MM. In the present studies, hexamethylene bisacetamide (HMBA), a hybrid polar compound that is a potent inducer of terminal differentiation of various transformed cells, is shown to inhibit the growth of several human myeloma cell lines (ARP-1, U266, and RPMI 8226), including doxorubicin-resistant RPMI 8226 variants that overexpress the multidrug-resistance gene, MDR-1, and its product, p-glycoprotein. In addition to growth arrest and suppression of clonogenicity, HMBA induces apoptosis both in freshly isolated human myeloma cells and in cell lines, as determined by morphologic alterations, cell cycle distribution and endonucleosomal DNA fragmentation. Further, HMBA decreases BCL-2 protein expression in myeloma cells within 12-48 hr. Overexpression of BCL-2 protein in ARP-1 cells confers resistance to HMBA-induced apoptosis. Taken together, these data suggest that HMBA is a potent inducer of apoptosis in human myeloma cells, which may act through suppressing the anti-apoptotic function of the bcl-2 gene. HMBA, and related hybrid polar compounds, may prove useful in the management of this presently incurable disease.


Subject(s)
Acetamides/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Down-Regulation , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Down-Regulation/drug effects , Humans , Immunoenzyme Techniques , Proto-Oncogene Proteins c-bcl-2/genetics , Transfection , Tumor Cells, Cultured
16.
Genomics ; 26(2): 327-33, 1995 Mar 20.
Article in English | MEDLINE | ID: mdl-7601459

ABSTRACT

The PML gene located on chromosome band 15q22 is involved with the RAR alpha locus (17q21) in a balanced reciprocal translocation uniquely observed in acute promyelocytic leukemia. Physical mapping studies by pulsed-field gel electrophoresis revealed that the PML gene is flanked by two CpG islands that are separated by a variable distance in normal individuals. Several lines of evidence demonstrate that this is the consequence of a large insertion/deletion polymorphism linked to the PML locus: (1) overlapping fragments obtained with a variety of rare-cutting restriction enzymes demonstrated the same variability in distance between the flanking CpG islands; (2) mapping with restriction enzymes insensitive to CpG methylation confirmed that the findings were not a consequence of variable methylation of CpG dinucleotides; (3) the polymorphism followed a Mendelian inheritance pattern. This polymorphism is localized 3' to the PML locus. There are five common alleles, described on the basis of BssHII fragments, ranging from 220 to 350 kb with increments of approximately 30 kb between alleles. Both heterozygous (61%) and homozygous (39%) patterns were observed in normal individuals. Megabase-scale insertion/deletion restriction fragment length polymorphisms are very rare and have been described initially in the context of multigene families. Such structures have been also reported as likely regions of genetic instability. High-resolution restriction mapping of this particular structure linked to the PML locus is underway.


Subject(s)
Chromosomes, Human, Pair 15 , Genes , Neoplasm Proteins , Nuclear Proteins , Polymorphism, Restriction Fragment Length , Transcription Factors/genetics , Alleles , Chromosomes, Human, Pair 17 , Deoxyribonucleases, Type II Site-Specific , Electrophoresis, Gel, Pulsed-Field , Genetic Linkage , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Promyelocytic Leukemia Protein , Receptors, Retinoic Acid/genetics , Reference Values , Repetitive Sequences, Nucleic Acid , Retinoic Acid Receptor alpha , Sequence Deletion , Translocation, Genetic , Tumor Cells, Cultured , Tumor Suppressor Proteins
17.
Br J Haematol ; 89(1): 217-8, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7833269

ABSTRACT

All-trans retinoic acid (ATRA) inhibits human myeloma cell growth in vitro, presumably through the down-regulation of interleukin 6 receptors (IL-6R). Based on these and other studies, we initiated a phase II clinical trial using ATRA in patients with advanced refractory multiple myeloma (MM). We report that three out of six treated patients developed severe hypercalcaemia following administration of ATRA, which was accompanied by a significant rise in serum IL-6 levels. Normal calcium levels were restored after the discontinuation of the drug and the administration of standard anti-hypercalcaemic care. We suspect that down-regulation of IL-6R resulted in increased serum IL-6 levels, leading to advanced bone resorption and hypercalcaemia. We conclude that the use of ATRA in patients with advanced MM is not warranted.


Subject(s)
Hypercalcemia/chemically induced , Interleukin-6/blood , Multiple Myeloma/drug therapy , Tretinoin/adverse effects , Humans , Multiple Myeloma/blood , Tretinoin/therapeutic use
18.
Genomics ; 11(4): 1054-62, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1686014

ABSTRACT

The mouse ob mutation has been mapped relative to a series of RFLPs among the progeny of three separate mouse crosses: an intraspecific backcross, an intraspecific intercross, and an interspecific intercross. Genotypic assignment at the ob locus was made by making use of measurements of body mass index and the plasma concentrations of glucose and insulin. These data have suggested that the development of diabetes in these animals is a consequence of unlinked polygenes. There was also evidence that unlinked Mus spretus alleles can diminish the obesity of ob/ob mice. From these data we have mapped several markers on chromosome 6 with the following order: cen-Cola-2-Met-ob-Cpa-Tcrb. The homologs of markers that flank ob map to human chromosome 7q, suggesting that if there is a human homologue of ob, it maps to 7q31.


Subject(s)
Mutation , Obesity/genetics , Animals , Blotting, Southern , Chromosome Mapping , Crosses, Genetic , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Mutant Strains , Pedigree , Phenotype , Polymorphism, Restriction Fragment Length
19.
Cell Immunol ; 101(2): 380-90, 1986 Sep.
Article in English | MEDLINE | ID: mdl-3093092

ABSTRACT

Interferon (IFN)-alpha and IFN-beta ("type I" IFNs), but not IFN-gamma reduced phytohemagglutinin- or pokeweed mitogen (PWM)-induced proliferation in cultures of human mononuclear leukocytes. Proliferation induced by specific antigens (tuberculin PPD or tetanus toxoid) or by exogenous interleukin 2 (IL-2) was strongly inhibited by type I IFNs and, to a lesser extent, by IFN-gamma as well. Inhibition of proliferation in mitogen-stimulated cultures was not due to a reduced production of IL-2 or to an inhibition of IL-2 receptor expression. Type I IFNs inhibited immunoglobulin (Ig) production in PWM-stimulated unseparated mononuclear cells, whereas IFN-gamma enhanced Ig production in such cultures. In cultures of purified B cells type I IFNs caused a stimulation of Ig production and this B-cell differentiation factor (BCDF)-like activity of IFNs was synergistically enhanced in the presence of IL-2. IFN-gamma produced less BCDF-like activity than type I IFNs. These results show that in some instances type I IFNs can be more potent in affecting functions of cells of the immune system than IFN-gamma.


Subject(s)
Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Antibody Formation/drug effects , Antigens, Differentiation, B-Lymphocyte , Antigens, Surface/biosynthesis , Cell Division/drug effects , Humans , Interleukin-2/pharmacology , Lectins/pharmacology , Lymphocytes/classification
20.
Cell Immunol ; 99(1): 287-93, 1986 Apr 15.
Article in English | MEDLINE | ID: mdl-3093100

ABSTRACT

The mechanism of human peripheral blood monocyte-mediated cytotoxicity was investigated using the HT-29 human colon adenocarcinoma line, A673 human rhabdomyosarcoma line, and A375 human melanoma line as target cells. Pretreatment of these target cells with 100 U/ml of recombinant human interferon (IFN)-gamma for 48 hr increased their susceptibility to monocyte killing. Increased susceptibility to the lytic action was particularly pronounced at low effector/target cell ratios. Unlike IFN-gamma human IFN-alpha did not potentiate monocyte cytotoxicity, and pretreatment of HT-29 with IFN-alpha also had virtually no effect on their susceptibility to monocyte killing. However, IFN-gamma appeared to prime either monocytes or target cells to become responsive to IFN-alpha. Our data suggest that IFN-gamma can promote the killing of tumor cells by monocytes through two separate actions, one on the monocyte and one on the target cell.


Subject(s)
Interferon-gamma/pharmacology , Monocytes/immunology , Adenocarcinoma , Cell Line , Colonic Neoplasms , Cytotoxicity, Immunologic/drug effects , Humans , Interferon Type I/pharmacology , Melanoma , Monocytes/drug effects , Recombinant Proteins/pharmacology , Rhabdomyosarcoma
SELECTION OF CITATIONS
SEARCH DETAIL
...