18 research outputs found

    17-β-Estradiol-dependent regulation of somatostatin receptor subtype expression in the 7315b prolactin secreting rat pituitary tumor in vitro and in vivo

    Get PDF
    In the present study, we have investigated the role of estrogens in the regulation of somatostatin receptor subtype (sst) expression in 7315b PRL- secreting rat pituitary tumor cells in vitro and in vivo. sst were undetectable in freshly dispersed cells of the transplantable 7315b tumor. When 7315b cells were cultured in medium containing 10% FCS, the number of high affinity sst increased with prolonged culture time. However, when the medium was supplemented with 10% horse serum (HS) instead of FCS, no sst were detectable on 7315b cells even after three weeks of culturing. In contrast to HS, FCS contains high E2-levels (HS, 8 pM; FCS, 134 pM). The antiestrogen tamoxifen (0.5 μM) significantly inhibited the sst number to 50.5% of the value of untreated FCS-grown cells, suggesting that E2 stimulates sst expression in 7315b rat pituitary tumor cells. E2 (l0 nM) induced a rapid increase in sst number in HS-grown 7315b cells. Octreotide (1μM) significantly inhibited PRL release and the intracellular PRL concentration of 7315b cells that were cultured in medium supplemented with FCS or with HS + l0 nM E2 but not in HS alone. This indicates that the sst present on these cells are biologically active. RT-PCR analysis revealed that none of the five currently known sst subtypes were present in freshly dispersed 7315b pituitary tumor cells. The expression of sst2- and sst3- messenger RNA (mRNA) was unequivocally correlated to the presence of E2 because these sst subtypes were detected only in cells that were cultured for7 and 14 days in medium supplemented with FCS or with HS + 10 nM E2. sst1, sst4 and sst5 messenger RNA could not be detected. The 7315b tumor itself synthesizes and secretes huge amounts of PRL. The high PRL levels in tumor-bearing rats inhibit the ovarian E2-production. No detectable E2 levels could be measured in the serum of 7315b tumor-bearing rats. The sc administration of 20 μg/day E2-benzoate normalized the circulating E2 levels in 7315b tumor- bearing rats. Moreover, E2-treatment indeed induced sst expression in vivo as shown by ligand binding studies using membrane homogenates and [125I- Tyr3]-octreotide as radioligand and by autoradiography on tissue sections. In agreement with the in vitro studies, the expression of the sst2 subtype was established by RT-PCR analysis in 7315b tumors of E2-treated rats. However, in contrast to the in vitro studies. E2-treatment did not effectuate the expression of the sst3 subtype, suggesting that the in vitro stimulus of E2 is stronger. In conclusion: 1) sst2 and sst3 expression in the 7315b rat prolactinoma model is primarily dependent upon the presence of estrogens; 2) the antihormonal action of octreotide in 7315b tumor cells in vitro is mediated via the sst2 and/or sst3 subtypes; 3) the absence of sst expression in vivo can be explained by the hormonal environment of the 7315b tumor cells. The 7315b tumor cells in vivo may down-regulate their own receptor status via their host, because of the ensuing hyperprolactinemia results in a hypo-estrogenic state.</p

    Determinants of lenalidomide response with or without erythropoiesis-stimulating agents in myelodysplastic syndromes:the HOVON89 trial

    Get PDF
    A randomized phase-II study was performed in low/int-1 risk MDS (IPSS) to study efficacy and safety of lenalidomide without (arm A) or with (arm B) ESA/G-CSF. In arm B, patients without erythroid response (HI-E) after 4 cycles received ESA; G-CSF was added if no HI-E was obtained by cycle 9. HI-E served as primary endpoint. Flow cytometry and next-generation sequencing were performed to identify predictors of response. The final evaluation comprised 184 patients; 84% non-del(5q), 16% isolated del(5q); median follow-up: 70.7 months. In arm A and B, 39 and 41% of patients achieved HI-E; median time-to-HI-E: 3.2 months for both arms, median duration of-HI-E: 9.8 months. HI-E was significantly lower in non-del(5q) vs. del(5q): 32% vs. 80%. The same accounted for transfusion independency-at-week 24 (16% vs. 67%), but similar in both arms. Apart from presence of del(5q), high percentages of bone marrow lymphocytes and progenitor B-cells, a low number of mutations, absence of ring sideroblasts, and SF3B1 mutations predicted HI-E. In conclusion, lenalidomide induced HI-E in patients with non-del(5q) and del(5q) MDS without additional effect of ESA/G-CSF. The identified predictors of response may guide application of lenalidomide in lower-risk MDS in the era of precision medicine. (EudraCT 2008-002195-10).</p

    Health-related quality of life in transplant ineligible newly diagnosed multiple myeloma patients treated with either thalidomide or lenalidomide-based regimen until progression: a prospective, open-label, multicenter, randomized, phase 3 study

    Get PDF
    Data on the impact of long term treatment with immunomodulatory drugs (IMiD) on health-related quality of life (HRQoL) is limited. The HOVON-87/NMSG18 study was a randomized, phase 3 study in newly diagnosed transplant ineligible patients with multiple myeloma, comparing melphalan-prednisolone in combination with thalidomide or lenalidomide, followed by maintenance therapy until progression (MPT-T or MPR-R). The EORTC QLQ-C30 and MY20 questionnaires were completed at baseline, after three and nine induction cycles and six and 12 months of maintenance therapy. Linear mixed models and minimal important differences were used for evaluation. 596 patients participated in HRQoL reporting. Patients reported clinically relevant improvement in global quality of life (QoL), future perspective and role and emotional functioning, and less fatigue and pain in both arms. The latter being of large effect size

    Somatostatin receptor manipulation

    No full text
    The expression of somatostatin receptors (ssts) on human tumours is the basis for the successful therapeutic and diagnostic application of (radiolabelled) somatostatin analogues. Manipulation (up-regulation) of sst expression might improve the uptake of radioligand in in vivo scintigraphy of human sst-positive tumours, as well as the potential success of radiotherapy using radiolabelled SRIF analogues. In clonal pituitary cell lines, agonist exposure (SRIF-14, SRIF-28, octreotide) has been shown to either reduce or increase sst (subtype) expression, suggesting cell-type-specific responsiveness. In addition, glucocorticoids and oestrogens were shown to down- and up-regulate, respectively, sst numbers. So far, little information is available with respect to sst (subtype) regulation in non-pituitary-derived cell types. We have found that sst expression in the model of the transplantable prolactin (PRL)-secreting rat pituitary tumour 7315b is mainly dependent upon the presence of oestradiol (E2), both in vivo and in vitro. This tumour is sst negative in vivo. In vitro, the addition of E2 induces sst expression (sst2 and sst3 subtypes). The in vivo administration of E2 (20 μg/day subcutaneously) to 7315b-tumour-bearing rats induces sst2 mRNA expression. The absence of sst expression in 7315b tumours in vivo may be due to the inhibition of ovarian E2 production by the high circulating PRL levels in the 7315b-prolactinoma-bearing rats. Indeed, no detectable E2 levels were found in the serum of 7315b-tumour-bearing rats. Taken together, our data suggest that the 7315b rat prolactinoma can indirectly manipulate (down-regulate) its own sst expression, in vivo, via its host. This experimental 7315b prolactinoma model might be representative for most untreated female prolactinoma patients. Clinically, patients with microprolactinomas do not benefit from octreotide treatment.</p

    Somatostatin receptor manipulation

    No full text
    The expression of somatostatin receptors (ssts) on human tumours is the basis for the successful therapeutic and diagnostic application of (radiolabelled) somatostatin analogues. Manipulation (up-regulation) of sst expression might improve the uptake of radioligand in in vivo scintigraphy of human sst-positive tumours, as well as the potential success of radiotherapy using radiolabelled SRIF analogues. In clonal pituitary cell lines, agonist exposure (SRIF-14, SRIF-28, octreotide) has been shown to either reduce or increase sst (subtype) expression, suggesting cell-type-specific responsiveness. In addition, glucocorticoids and oestrogens were shown to down- and up-regulate, respectively, sst numbers. So far, little information is available with respect to sst (subtype) regulation in non-pituitary-derived cell types. We have found that sst expression in the model of the transplantable prolactin (PRL)-secreting rat pituitary tumour 7315b is mainly dependent upon the presence of oestradiol (E2), both in vivo and in vitro. This tumour is sst negative in vivo. In vitro, the addition of E2 induces sst expression (sst2 and sst3 subtypes). The in vivo administration of E2 (20 μg/day subcutaneously) to 7315b-tumour-bearing rats induces sst2 mRNA expression. The absence of sst expression in 7315b tumours in vivo may be due to the inhibition of ovarian E2 production by the high circulating PRL levels in the 7315b-prolactinoma-bearing rats. Indeed, no detectable E2 levels were found in the serum of 7315b-tumour-bearing rats. Taken together, our data suggest that the 7315b rat prolactinoma can indirectly manipulate (down-regulate) its own sst expression, in vivo, via its host. This experimental 7315b prolactinoma model might be representative for most untreated female prolactinoma patients. Clinically, patients with microprolactinomas do not benefit from octreotide treatment.</p

    Distinct bone marrow immunophenotypic features define the splicing factor 3B subunit 1 (SF3B1)-mutant myelodysplastic syndromes subtype

    No full text
    Splicing factor 3B subunit 1 (SF3B1) mutations define a distinct myelodysplastic syndromes (MDS) patient group with a relatively favourable disease course and high response rates to luspatercept. Few data are available on bone marrow phenotype beyond ring sideroblasts in this subgroup of patients with MDS. In the present study, we identified immunophenotypic erythroid, myelomonocyte and progenitor features associated with SF3B1 mutations. In addition, we illustrate that SF3B1-mutation type is associated with distinct immunophenotypic features, and show the impact of co-occurrence of a SF3B1 mutation and a deletion of chromosome 5q on bone marrow immunophenotype. These genotype–phenotype associations and phenotypic subtypes within SF3B1-MDS provide leads that may further refine prognostication and therapeutic strategies for this particular MDS subgroup
    corecore