Interdisziplinäre Empfehlungen zur Behandlung des metastasierten Nierenzellkarzinoms

2017 ◽  
Vol 38 (06) ◽  
pp. 373-380
Author(s):  
Kurt Miller ◽  
Lothar Bergmann ◽  
Christian Doehn ◽  
Jürgen Gschwend ◽  
Ulrich Keilholz

ZusammenfassungDie Prognose von Patienten mit einem metastasierten Nierenzellkarzinom (mRCC) hat sich dank zielgerichteter Substanzen deutlich verbessert. Überlebenszeiten von mehr als 2 Jahren sind realistisch. Die Fortschritte zeigen sich auch darin, dass zunehmend Therapieoptionen in Dritt- und weiteren Linien thematisiert werden.Sunitinib, Pazopanib, die Kombination Bevacizumab+Interferon-alpha sowie Temsirolimus sind für die Erstlinie beim mRCC zugelassen. Sunitinib und Pazopanib haben eine Zulassung auch für die Zweitlinientherapie – für Pazopanib ist diese auf den Einsatz nach Zytokinen beschränkt. Everolimus (nach Therapie mit einem Tyrosinkinase-Inhibitor; TKI), Axitinib (nach Sunitinib- oder Zytokintherapie) sowie Sorafenib (nach Zytokintherapie) sind weitere Wirkstoffe für die Zweitlinien-Therapie.In der Zweitlinientherapie gab es 3 Neuzulassungen: Nach Versagen einer gegen VEGF gerichteten Therapie können nun Nivolumab, Cabozantinib und Lenvatinib in Kombination mit Everolimus eingesetzt werden. Damit stehen zum ersten Mal eine zielgerichtete Immuntherapie und eine Kombination zielgerichteter Substanzen beim mRCC zur Verfügung.Zur Frage nach der optimalen Sequenztherapie gibt es keine neuen Erkenntnisse. Bisher liegen Ergebnisse einer Phase-III-Studie vor, in der die geprüften Sequenzen Sorafenib-Sunitinib und Sequenz Sunitinib-Sorafenib gleichwertig waren.Ziel eines interdisziplinären RCC-Expertengesprächs war es, gemeinsame Therapieempfehlungen auf Basis der aktuell publizierten Daten und der eigenen klinischen Erfahrung zu erarbeiten und für den Praxisalltag abzuleiten. Die Ergebnisse werden in dieser Publikation vorgestellt.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3177-3177
Author(s):  
Heinz Gisslinger ◽  
Veronika Buxhofer-Ausch ◽  
Josef Thaler ◽  
Ernst Schloegl ◽  
Guenther A. Gastl ◽  
...  

Abstract Background AOP2014 is a next generation long-acting pegylated IFN-alpha-2b, consisting predominantly of only one isoform, as opposed to other commercially available pegylated interferons. Due to this property, reduced dosing frequencies, better tolerability, improved compliance and more favorable long-term treatment outcomes in patients with Polycythemia Vera (PV) are expected. The drug has Orphan designation by EMA and FDA and is currently in the phase III stage of development. Study design This phase I/II single arm dose escalation study with cohort extension included 51 patients with PV who could be either cytoreduction therapy naive or pre-treated. AOP2014 was administered subcutaneously in a dose range of 50-540 mcg every two weeks. Main objectives were to define the maximum tolerated dose as well as observe the long term safety and efficacy in terms of normalization of blood parameters and molecular abnormalities (results were already presented at the ASH 2013 annual meeting by Gisslinger et al). The option to switch to an "once every four weeks" schedule has been implemented by the amended protocol for patients who responded well to the treatment and participated in the study for longer than one year. The dose of the study drug had to remain unchanged after the switch, resulting in a decrease (by approx. the half) of the overall exposure to the drug. Outcomes of this switch are presented here. Results Patients (period A, median exposure duration 34 weeks, mean monthly dose 484 mcg) were dosed every two weeks based on the Phase II dosing rules prior having switch option. 33 patients (period B, median exposure duration 12 weeks, mean monthly dose 413 mcg) were dosed every two weeks beyond the first year and, showing benefit from treatment, have been assessed as eligible for switch. 28 patients (period C, median exposure duration 42 weeks) were then switched to once every four weeks schedule (mean monthly dose 221 mcg). Blood parameters were normalized and remained stable following one year of treatment and could be maintained after the switch (hematocrit, median,% - period A: 43, period B: 43, period C: 42; WBC, median, G/l - period A: 6.1, period B: 5.9, period C: 5.7; platelets, median, G/l - period A: 246, period B: 211, period C: 204). Spleen length stayed stable within the normal range following the switch in the majority of patients either (mean, in cm – period A: 11.4, period B: 8.3, period C: 10.3). Complete response as best individual response could be maintained in 42% from the period A, 55% in the period B and 67% of the period C patients, while for the partial hematological responders the results were 60%, 71% and 67%, respectively. Molecular response improved continuously over time, being maintained at the best individual level in 31% of period A patients, compared to 42% of period B and 75% of period C patients. Decrease of application frequency and total dose exposure led to decrease of the occurrence of all/drug related AEs (measured as mean count of adverse events [AE] per patient week exposure) to 0.17/0.09 (arm A) from 0.3/0.09 (arm B) and 0.08/0.03 (arm C). Conclusions This explorative data from endpoints pre-defined in the prospective study demonstrate the feasibility to further reduce the frequency of AOP2014 administration to once every four weeks in responding patients, previously treated every two weeks. Reduced injection frequency is not associated with a lack of- response, but clearly improves tolerability. Finally, continuous reduction of the JAK2 allelic burden indicates that duration of interferon exposure rather than dose of interferon is an important variable inducing molecular responses. The here presented findings support the idea that interferon alpha effects in PV are pleiotropic, such as induction of immune-surveillance, which is continuously maintained at lower AOP2014 levels. Disclosures Gisslinger: AOP Orphan Pharmaceuticals AG: Research Funding. Buxhofer-Ausch:AOP Orphan Pharmaceuticals AG: Research Funding. Thaler:AOP Orphan Pharmaceuticals AG: Research Funding. Schloegl:AOP Orphan Pharmaceuticals AG: Research Funding. Gastl:AOP Orphan Pharmaceuticals AG: Research Funding. Wolf:AOP Orphan Pharmaceuticals AG: Research Funding. Kralovics:AOP Orphan Pharmaceuticals AG: Research Funding. Gisslinger:AOP Orphan Pharmaceuticals AG: Research Funding. Strecker:AOP Orphan Pharmaceuticals AG: Research Funding. Egle:AOP Orphan Pharmaceuticals AG: Research Funding. Melchardt:AOP Orphan Pharmaceuticals AG: Research Funding. Burgstaller:AOP Orphan Pharmaceuticals AG: Research Funding. Willenbacher:AOP Orphan Pharmaceuticals AG: Research Funding. Zoerer:AOP Orphan Pharmaceuticals AG: Employment. Kadlecova:AOP Orphan Pharmaceuticals AG: Consultancy. Zagrijtschuk:AOP Orphan Pharmaceuticals AG: Employment. Klade:AOP Orphan Pharmaceuticals AG: Employment. Greil:AOP Orphan Pharmaceuticals AG: Research Funding.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 424-424 ◽  
Author(s):  
Hartmut Goldschmidt ◽  
Pieter Sonneveld ◽  
Iris Breitkreuz ◽  
Bronno van der Holt ◽  
Axel Benner ◽  
...  

Abstract The joint phase-III HOVON50/GMMG-HD3 trial was designed to assess the effect of thalidomide in induction treatment and as maintenance after high-dose therapy (HDT) and autologous stem cell transplantation (SCT) for multiple myeloma (MM). The standard treatment arm comprised 3 cycles of VAD, mobilisation with CAD+G-CSF (cyclophosphamide 1000 mg/m2, day 1; adriamycin 15 mg/m2, days 1–4; dexamethasone 40 mg, days 1–4; G-CSF until end of harvest), HDT with 1 or 2 cycles of melphalan 200 mg/m2, followed by autologous peripheral blood SCT (PBSCT), and maintenance with interferon-alpha (9 mio. U per week). In the experimental arm, TAD (thalidomide, 200 mg for HOVON / 400 mg for GMMG; adriamycin 9 mg/m2, days 1–4; dexamethasone 40 mg, days 1–4, 9–12, 17–21) was used for induction treatment. Mobilisation and HDT were identical to the standard arm. Experimental maintenance was thalidomide (50 mg per day). A first group of 406 patients (of 1050 included) are evaluable for the comparison of VAD vs. TAD and response after 1st HDT. A trend for a higher toxicity was observed in the TAD- compared with the VAD-arm (drop out: 15% vs. 8%, p= 0.10). Low molecular weight heparin was effective in the prevention of deep venous thrombosis during TAD-treatment (DVT-incidence 8% vs. 4% p= 0.15). The median number of stem cell collections to harvest at least one autograft was 1 in both arms (p= n.s.). Treatment-results are presented in table 1. In a subgroup of 90 GMMG-patients, 78% and 74% compared with 62% and 54% still received thalidomide compared with interferon-alpha at 12 and 24 months after start of maintenance. In summary, thalidomide+AD induce a significantly higher response rate, but this effect is completely offset by HDM. Therefore, results on EFS/PFS are necessary before thalidomide containing regimens can be defined as a standard for induction treatment before HDT. The maintenance treatment with thalidomide is better tolerated compared with interferon-alpha. Treatment results (n=406) after VAD/TAD and after first HDT After VAD After TAD p-value PR 60% 73% <0.001 CR 3% 7% 0.11 PR/CR 63% 80% 0.001 After VAD/HDT After TAD/HDT p-value PR 75% 72% 0.8 CR 13% 19% 0.3 PR/CR 88% 91% 0.4


Sign in / Sign up

Export Citation Format

Share Document