Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Janusz Milanowski is active.

Publication


Featured researches published by Janusz Milanowski.


Journal of Clinical Oncology | 2007

Phase III Study of Erlotinib in Combination With Cisplatin and Gemcitabine in Advanced Non–Small-Cell Lung Cancer: The Tarceva Lung Cancer Investigation Trial

Ulrich Gatzemeier; Anna Pluzanska; Aleksandra Szczesna; E. Kaukel; Jaromír Roubec; Flavio De Rosa; Janusz Milanowski; Hanna Karnicka-Mlodkowski; Miloš Pešek; Piotr Serwatowski; Rodryg Ramlau; Terezie Janaskova; Johan Vansteenkiste; Janos Strausz; Georgy M. Manikhas; Joachim von Pawel

PURPOSE Erlotinib is a potent inhibitor of the epidermal growth factor receptor tyrosine kinase, with single-agent antitumor activity. Preclinically, erlotinib enhanced the cytotoxicity of chemotherapy. This phase III, randomized, double-blind, placebo-controlled, multicenter trial evaluated the efficacy and safety of erlotinib in combination with cisplatin and gemcitabine as first-line treatment for advanced non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS Patients received erlotinib (150 mg/d) or placebo, combined with up to six 21-day cycles of chemotherapy (gemcitabine 1,250 mg/m2 on days 1 and 8 and cisplatin 80 mg/m2 on day 1). The primary end point was overall survival (OS). Secondary end points included time to disease progression (TTP), response rate (RR), duration of response, and quality of life (QoL). RESULTS A total of 1,172 patients were enrolled. Baseline demographic and disease characteristics were well balanced. There were no differences in OS (hazard ratio, 1.06; median, 43 v 44.1 weeks for erlotinib and placebo groups, respectively), TTP, RR, or QoL between treatment arms. In a small group of patients who had never smoked, OS and progression-free survival were increased in the erlotinib group; no other subgroups were found more likely to benefit. Erlotinib with chemotherapy was generally well tolerated; incidence of adverse events was similar between arms, except for an increase in rash and diarrhea with erlotinib (generally mild). CONCLUSION Erlotinib with concurrent cisplatin and gemcitabine showed no survival benefit compared with chemotherapy alone in patients with chemotherapy-naïve advanced NSCLC.


Annals of Oncology | 2015

Randomized, phase III trial of figitumumab in combination with erlotinib versus erlotinib alone in patients with nonadenocarcinoma nonsmall-cell lung cancer

Giorgio V. Scagliotti; Igor Bondarenko; Fiona Blackhall; Fabrice Barlesi; Te-Chun Hsia; Jacek Jassem; Janusz Milanowski; Sanjay Popat; J. M. Sanchez-Torres; Silvia Novello; Rebecca J. Benner; S. Green; K. Molpus; Frances A. Shepherd

BACKGROUND Figitumumab (CP-751,871) is a fully human IgG2 monoclonal antibody that inhibits the insulin-like growth factor 1 receptor. This multicenter, randomized, phase III study investigated the efficacy of figitumumab plus erlotinib compared with erlotinib alone in patients with pretreated, nonsmall-cell lung cancer (NSCLC). PATIENTS AND METHODS Patients (stage IIIB/IV or recurrent disease with nonadenocarcinoma histology) who had previously received at least one platinum-based regimen were randomized to receive open-label figitumumab (20 mg/kg) plus erlotinib 150 mg/day or erlotinib alone every 3 weeks. The primary end point was overall survival (OS). RESULTS Of 583 patients randomized, 579 received treatment. The study was closed early by an independent data safety monitoring committee due to results crossing the prespecified futility boundary. At the final analysis, median OS was 5.7 months for figitumumab plus erlotinib and 6.2 months for erlotinib alone [hazard ratio (HR) 1.09; 95% confidence interval (CI) 0.91-1.31; P = 0.35]. Median progression-free survival was 2.1 months for figitumumab plus erlotinib and 2.6 months for erlotinib alone (HR 1.08; 95% CI 0.90-1.29; P = 0.43). Treatment-related nonfatal serious adverse events occurred in 18% and 5% of patients in the figitumumab arm or erlotinib alone arm, respectively. There were nine treatment-related deaths (three related to both drugs, four related to erlotinib alone and two related to figitumumab). CONCLUSIONS The addition of figitumumab to erlotinib did not improve OS in patients with advanced, pretreated, nonadenocarcinoma NSCLC. Clinical development of figitumumab has been discontinued. CLINICAL TRIAL ID NCT00673049.


Annals of Oncology | 2015

Cilengitide combined with cetuximab and platinum-based chemotherapy as first-line treatment in advanced non-small-cell lung cancer (NSCLC) patients: results of an open-label, randomized, controlled phase II study (CERTO)

Johan Vansteenkiste; Fabrice Barlesi; C. Waller; Jaafar Bennouna; C. Gridelli; Eray Goekkurt; Dennis Verhoeven; Aleksandra Szczesna; M. Feurer; Janusz Milanowski; Paul Germonpre; H. Lena; Djordje Atanackovic; Maciej Krzakowski; C. Hicking; J. Straub; M. Picard; Wolfgang Schuette; Kenneth J. O'Byrne

BACKGROUND This multicentre, open-label, randomized, controlled phase II study evaluated cilengitide in combination with cetuximab and platinum-based chemotherapy, compared with cetuximab and chemotherapy alone, as first-line treatment of patients with advanced non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS Patients were randomized 1:1:1 to receive cetuximab plus platinum-based chemotherapy alone (control), or combined with cilengitide 2000 mg 1×/week i.v. (CIL-once) or 2×/week i.v. (CIL-twice). A protocol amendment limited enrolment to patients with epidermal growth factor receptor (EGFR) histoscore ≥200 and closed the CIL-twice arm for practical feasibility issues. Primary end point was progression-free survival (PFS; independent read); secondary end points included overall survival (OS), safety, and biomarker analyses. A comparison between the CIL-once and control arms is reported, both for the total cohorts, as well as for patients with EGFR histoscore ≥200. RESULTS There were 85 patients in the CIL-once group and 84 in the control group. The PFS (independent read) was 6.2 versus 5.0 months for CIL-once versus control [hazard ratio (HR) 0.72; P = 0.085]; for patients with EGFR histoscore ≥200, PFS was 6.8 versus 5.6 months, respectively (HR 0.57; P = 0.0446). Median OS was 13.6 for CIL-once versus 9.7 months for control (HR 0.81; P = 0.265). In patients with EGFR ≥200, OS was 13.2 versus 11.8 months, respectively (HR 0.95; P = 0.855). No major differences in adverse events between CIL-once and control were reported; nausea (59% versus 56%, respectively) and neutropenia (54% versus 46%, respectively) were the most frequent. There was no increased incidence of thromboembolic events or haemorrhage in cilengitide-treated patients. αvβ3 and αvβ5 expression was neither a predictive nor a prognostic indicator. CONCLUSIONS The addition of cilengitide to cetuximab/chemotherapy indicated potential clinical activity, with a trend for PFS difference in the independent-read analysis. However, the observed inconsistencies across end points suggest additional investigations are required to substantiate a potential role of other integrin inhibitors in NSCLC treatment. CLINICAL TRIAL REGISTRATION ID NUMBER NCT00842712.


Journal of Thoracic Oncology | 2008

Phase I/II Study of a 3 Weekly Oral Taxane (DJ-927) in Patients with Recurrent, Advanced Non-small Cell Lung Cancer

Paul Baas; Aleksandra Szczesna; Istvan Albert; Janusz Milanowski; Erzsébet Juhász; Z. Sztancsik; J von Pawel; R. Oyama; Sjaak Burgers

Introduction: A phase I/II study was performed to assess the efficacy and toxicity of a new oral taxane in patients with recurrent, advanced Non-small Cell Lung Cancer. Patients and Methods: Patients who were treated with one prior, taxane free chemotherapy regimen, were eligible for this study. A single oral dose of DJ-927 (27 mg/m2) was given every 3 weeks. In case of good tolerance, one dose escalation to 35 mg/m2 was allowed. Response and toxicity were measured and plasma pharmacokinetic analysis was performed during the first course. Results: From October 2004 to September 2005, 36 patients gave informed consent and 34 received medication. The mean age was 58 years (range, 33–75 years). The majority of patients were pretreated with a combination of cisplatin and gemcitabine. Median interval between end of first treatment and the registration of this study was 7 months (range, 0.8–22 months). Twelve patients died on study of which eight due to disease progression. In four patients with preexisting cardiac disease, toxicity led to cardiac worsening and subsequent death. Grade 3 and 4 toxicities according to the National Cancer Institute Common Toxicity Criteria were neutropenia in 18 patients (53%), anemia in six patients (18%), nausea and fatigue in two patients (6%), febrile neutropenia and neurotoxicity in one patient (3%). The overall response rate for all patients was 5.6% (Confidence Interval [CI] 0.7–18.7%). The percentage of patients with stabilization for >6 weeks was 47%. The median time to progression was 97 days (CI: 47–167 days) and the median survival time was 120 days (CI: 68–222 days) for the ITT group. Since only a minority of patients (3) tolerated the higher drug dose we omitted this dose level because of hematological toxicity. Pharmacokinetic analysis showed that the median area under the curve (t = 0–168 hours) was 1752 ± 1355 ngr/ml/h and the half-life was 167 ± 77 hours. Conclusion: When administered once every 3 weeks, this oral taxane formulation of DJ-927 was well-absorbed with a long terminal half-life of 167 ± 77 hour. DJ-927 has antitumor activity against Non-small Cell Lung Cancer when given as second-line monotherapy (overall response rate in 5.6%; CI 0.7–18.7%). Ten patients experienced SD for more than 8 weeks. Different types of dose administration (metronomic dosing) or combination with other cytotoxic agents should be considered in future studies.


Folia Histochemica Et Cytobiologica | 2012

Predictive value of ERCC1 single-nucleotide polymorphism in patients receiving platinum-based chemotherapy for locally-advanced and advanced non-small cell lung cancer — a pilot study

Paweł Krawczyk; Kamila Wojas-Krawczyk; Radosław Mlak; Tomasz Kucharczyk; Beata Biernacka; Janusz Milanowski

Platinum-based chemotherapy is the main type of I-line treatment of advanced and non-operative NSCLC patients without EGFR gene mutation. The excision repair cross-complementation group 1 (ERCC1) is an enzyme that executes the incision of the damaged DNA strand and removes platinum-induced DNA adducts. We investigated whether ERCC1 gene polymorphism has an effect on the response to chemotherapy and survival in 43 patients with NSCLC treated with platinum-based chemotherapy. ERCC1 19007 T>C SNPs were assessed using a PCR-RFLP methods in DNA isolated from peripheral blood lymphocytes. Disease control occurred significantly (p = 0.045) more frequently in patients with CC or CT genotype compared to patients with TT genotype. Median PFS and OS for CC homozygous were 4 and 10.5 months, 4 and 12.5 months for CT heterozygous, but only 0.3 and 1.5 months for TT homozygous patients, respectively. The probability of PFS was significantly higher (HR = 0.438, 95% CI: 0.084–0.881, p = 0.03) and probability of OS was insignificantly higher (HR = 0.503, 95% CI: 0.129–1.137, p = 0.084) in patients with CC or CT genotype than in patients with TT genotype. Uncommon TT genotype of ERCC1 19007 T>C polymorphism could predict poor response and shortening of progression free survival in NSCLC patients treated with platinum-based I-line chemotherapy. The analysis of this polymorphism may serve as a promising tool in the qualification of advanced NSCLC patients for appropriate chemotherapy.


Chemotherapy | 2012

The Qualification of Docetaxel or Erlotinib for Second-Line Therapy Should Be Based on Clinical and Molecular Predictive Factors

Paweł Krawczyk; Dariusz M. Kowalski; Kamila Wojas-Krawczyk; Radosław Mlak; Piotr Jaśkiewicz; Tomasz Kucharczyk; Kinga Winiarczyk; Maciej Krzakowski; Janusz Milanowski

Background: We evaluated the effectiveness of docetaxel or erlotinib in second-line treatment of non-small cell lung cancer (NSCLC) and focused on the impact of predictive factors on the outcome of therapy. Methods: 204 patients with progressive disease after platinum-based therapy were enrolled: 102 received an infusion of 75 mg/m2 of docetaxel and 102 received 150 mg of erlotinib orally. Results: Response rate (RR) was 6.9 and 8.8% for docetaxel and erlotinib, respectively. Progression-free survival (PFS) was 1.2 months for docetaxel and 1.6 months for erlotinib (hazard ratio, HR = 1.2, p = 0.17). Overall survival was 5.5 versus 7 months for docetaxel and erlotinib, respectively (HR = 1.35, p = 0.06). Using Cox regression, we found clinical factors (performance status and weight loss) with predictive values for RR and PFS in second-line-treated patients. Prior radiotherapy, smoking status and EGFR mutation might help to predict outcome of erlotinib treatment and βIII-tubulin mRNA expression that of docetaxel, but histopathological diagnosis did not have any predictive value. Conclusions: Erlotinib and docetaxel show similar efficacy in the treatment of NSCLC. The application of predictive factors may facilitate qualification for second-line treatment with both drugs.


Annals of Oncology | 2017

ROMANA 3: a phase 3 safety extension study of anamorelin in advanced non-small-cell lung cancer (NSCLC) patients with cachexia

Jennifer S. Temel; Amy P. Abernethy; Janusz Milanowski; John Friend; Kenneth Fearon

Abstract Background Cancer anorexia–cachexia is a debilitating condition frequently observed in NSCLC patients, characterized by decreased body weight, reduced food intake, and impaired quality of life. Anamorelin, a novel selective ghrelin receptor agonist, has anabolic and appetite-enhancing activities. Patients and methods ROMANA 3 was a safety extension study of two phase 3, double-blind studies that assessed safety and efficacy of anamorelin in advanced NSCLC patients with cachexia. Patients with preserved Eastern Cooperative Oncology Group ≤2 after completing 12 weeks (w) on the ROMANA 1 or ROMANA 2 trials (0–12 weeks) could enroll in ROMANA 3 and continue to receive anamorelin 100 mg or placebo once daily for an additional 12w (12–24 weeks). The primary endpoint of ROMANA 3 was anamorelin safety/tolerability (12–24 weeks). Secondary endpoints included changes in body weight, handgrip strength (HGS), and symptom burden (0–24 weeks). Results Of the 703 patients who completed ROMANA 1 and ROMANA 2, 513 patients entered ROMANA 3 (anamorelin, N = 345, mean age 62.0 years; placebo, N = 168; mean age 62.2 years). During ROMANA 3, anamorelin and placebo groups had similar incidences of treatment–emergent adverse events (TEAEs; 52.2% versus 55.7%), grade ≥3 TEAEs (22.4% versus 21.6%), and serious TEAEs (12.8% versus 12.6%). There were 36 (10.5%) and 23 (13.8%) deaths in the anamorelin and placebo groups, respectively; none were drug-related. Improvements in body weight and anorexia–cachexia symptoms observed in the original trials were consistently maintained over 12–24 weeks. Anamorelin, versus placebo, significantly increased body weight from baseline of original trials at all time points (P < 0.0001) and improved anorexia–cachexia symptoms at weeks 3, 6, 9, 12, and 16 (P < 0.05). No significant improvement in HGS was seen in either group. Conclusion During the 12–24 weeks ROMANA 3 trial, anamorelin continued to be well tolerated. Over the entire 0–24w treatment period, body weight and symptom burden were improved with anamorelin. Clinical trial registration numbers ROMANA 1 (NCT01387269), ROMANA 2 (NCT01387282), and ROMANA 3 (NCT01395914).


Oncology Reports | 2013

Predictive value of ERCC1 and RRM1 gene single-nucleotide polymorphisms for first-line platinum- and gemcitabine-based chemotherapy in non-small cell lung cancer patients

Radosław Mlak; Paweł Krawczyk; Rodryg Ramlau; Ewa Kalinka-Warzocha; Maja Wasylecka-Morawiec; Kamila Wojas-Krawczyk; Tomasz Kucharczyk; Iwona Homa; Piotr Kozioł; Marzanna Ciesielka; Dorota Chudziak; Janusz Milanowski

Platinum-based chemotherapy with third generation drugs (such as gemcitabine) is an efficacious regimen of first-line treatment of patients with advanced, unresectable non-small cell lung cancer (NSCLC), without activating EGFR mutations. Mechanism of action of cytostatics are distortions in the DNA. ERCC1 and RRM1 are key proteins involved in the repair of DNA, thus, they may be responsible for the ineffectiveness of therapy. We investigated whether ERCC1 (19007C>T) and RRM1 (-37C>A) polymorphisms impact response to chemotherapy and survival in 62 patients with NSCLC treated with platinum and gemcitabine. Single nucleotide polymorphisms (SNPs) were assessed using a PCR-RFLP method in DNA isolated from PBLs. There were no statistically significant relationships between ERCC1 genotypes and response to therapy (p=0.581, χ2=1.09) as well as patient overall survival (OS). Carriers of the RRM1 AC genotype showed disease progression significantly more frequently (p=0.019, χ2=5.473) compared to carriers of the AA or CC genotypes. Carriers of the ERCC1/RRM1TT/CC genotype combination showed disease control significantly more frequently (p=0.047, χ2=3.95) compared to carriers of other genotype combinations. Patients with AA or CC genotypes of RRM1 showed significantly higher progression-free survival probability (p=0.0001, HR=0.39, 95% CI, 0.22-0.70) and OS probability (p=0.0104, HR=0.39, 95% CI, 0.18-0.82) compared to those with the AC genotype. In Cox regression model, poor performance status (p=0.0016, HR=4.78, 95% CI, 1.82-12.56), AC genotype of RRM1 gene (p=0.0414, HR=2.47, 95% CI, 1.04-5.87), lack of prior surgical treatment (p=0.0425, HR=4.71, 95% CI, 1.06-20.92) and lack of subsequent lines of treatment (p=0.0127, HR=3.23, 95% CI, 1.29-8.11) were significantly associated with shortening of patient survival. The analysis of RRM1 (-37C>A) more than ERCC1 (19007C>T) polymorphism may be a promising tool in the qualification of NSCLC patients for chemotherapy containing platinum compounds and gemcitabine.


Journal of Cancer Research and Clinical Oncology | 2014

Polymorphisms in TS, MTHFR and ERCC1 genes as predictive markers in first‑line platinum and pemetrexed therapy in NSCLC patients

Paweł Krawczyk; Tomasz Kucharczyk; Dariusz M. Kowalski; Tomasz Powrózek; Rodryg Ramlau; Ewa Kalinka-Warzocha; Kinga Winiarczyk; Magdalena Knetki-Wróblewska; Kamila Wojas-Krawczyk; Katarzyna Kałakucka; Wojciech Dyszkiewicz; Maciej Krzakowski; Janusz Milanowski

AbstractPurpose We presented retrospective analysis of up to five polymorphisms in TS, MTHFR and ERCC1 genes as molecular predictive markers for homogeneous Caucasian, non-squamous NSCLC patients treated with pemetrexed and platinum front-line chemotherapy.MethodsThe following polymorphisms in DNA isolated from 115 patients were analyzed: various number of 28-bp tandem repeats in 5′-UTR region of TS gene, single nucleotide polymorphism (SNP) within the second tandem repeat of TS gene (G>C); 6-bp deletion in 3′-UTR region of the TS (1494del6); 677C>T SNP in MTHFR; 19007C>T SNP in ERCC1. Molecular examinations’ results were correlated with disease control rate, progression-free survival (PFS) and overall survival.ResultsPolymorphic tandem repeat sequence (2R, 3R) in the enhancer region of TS gene and G>C SNP within the second repeat of 3R allele seem to be important for the effectiveness of platinum and pemetrexed in first-line chemotherapy. The insignificant shortening of PFS in 3R/3R homozygotes as compared to 2R/2R and 2R/3R genotypes were observed, while it was significantly shorter in patients carrying synchronous 3R allele and G nucleotide. The combined analysis of TS VNTR and MTHFR 677C>T SNP revealed shortening of PFS in synchronous carriers of 3R allele in TS and two C alleles in MTHFR. The strongest factors increased the risk of progression were poor PS, weight loss, anemia and synchronous presence of 3R allele and G nucleotide in the second repeat of 3R allele in TS. Moreover, lack of application of second-line chemotherapy, weight loss and poor performance status and above-mentioned genotype of TS gene increased risk of early mortality.ConclusionThe examined polymorphisms should be accounted as molecular predictor factors for pemetrexed- and platinum-based front-line chemotherapy in non-squamous NSCLC patients.


Annals of Agricultural and Environmental Medicine | 2016

Pantoea agglomerans : a mysterious bacterium of evil and good. Part III. Deleterious effects: infections of humans, animals and plants

Jacek Dutkiewicz; Barbara Mackiewicz; Marta Kinga Lemieszek; Marcin Golec; Janusz Milanowski

Pantoea agglomerans, a bacterium associated with plants, is not an obligate infectious agent in humans. However, it could be a cause of opportunistic human infections, mostly by wound infection with plant material, or as a hospital-acquired infection, mostly in immunocompromised individuals. Wound infection with P. agglomerans usually follow piercing or laceration of skin with a plant thorn, wooden splinter or other plant material and subsequent inoculation of the plant-residing bacteria, mostly during performing of agricultural occupations and gardening, or children playing. Septic arthritis or synovitis appears as a common clinical outcome of exogenous infection with P. agglomerans, others include endophthalmitis, periostitis, endocarditis and osteomyelitis. Another major reason for clinical infection with P. agglomerans is exposure of hospitalized, often immunodeficient individuals to medical equipment or fluids contaminated with this bacterium. Epidemics of nosocomial septicemia with fatal cases have been described in several countries, both in adult and paediatric patients. In most cases, however, the clinical course of the hospital-acquired disease was mild and application of the proper antibiotic treatment led to full recovery. Compared to humans, there are only few reports on infectious diseases caused by Pantoea agglomerans in vertebrate animals. This species has been identified as a possible cause of equine abortion and placentitis and a haemorrhagic disease in dolphin fish (Coryphaena hippurus). P. agglomerans strains occur commonly, usually as symbionts, in insects and other arthropods. Pantoea agglomerans usually occurs in plants as an epi- or endophytic symbiont, often as mutualist. Nevertheless, this species has also also been identified as a cause of diseases in a range of cultivable plants, such as cotton, sweet onion, rice, maize, sorghum, bamboo, walnut, an ornamental plant called Chinese taro (Alocasia cucullata), and a grass called onion couch (Arrhenatherum elatius). Some plant-pathogenic strains of P. agglomerans are tumourigenic, inducing gall formation on table beet, an ornamental plant gypsophila (Gypsophila paniculata), wisteria, Douglas-fir and cranberry. Recently, a Pantoea species closely related to P. agglomerans has been identified as a cause of bacterial blight disease in the edible mushroom Pleurotus eryngii cultivated in China. The genetically governed determinants of plant pathogenicity in Pantoea agglomerans include such mechanisms as the hypersensitive response and pathogenicity (hrp) system, phytohormones, the quorum-sensing (QS) feedback system and type III secretion system (T3SS) injecting the effector proteins into the cytosol of a plant cell.

Collaboration


Dive into the Janusz Milanowski's collaboration.

Top Co-Authors

Avatar

Paweł Krawczyk

Medical University of Lublin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomasz Powrózek

Medical University of Lublin

View shared research outputs
Top Co-Authors

Avatar

Marcin Nicoś

Medical University of Lublin

View shared research outputs
Top Co-Authors

Avatar

Tomasz Kucharczyk

Medical University of Lublin

View shared research outputs
Top Co-Authors

Avatar

Bożena Jarosz

Medical University of Lublin

View shared research outputs
Top Co-Authors

Avatar

Radosław Mlak

Medical University of Lublin

View shared research outputs
Top Co-Authors

Avatar

Barbara Mackiewicz

Medical University of Lublin

View shared research outputs
Top Co-Authors

Avatar

Rodryg Ramlau

Poznan University of Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Jacek Dutkiewicz

National Institute for Occupational Safety and Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge