Leo Rasche
University of Arkansas for Medical Sciences
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Leo Rasche.
European Journal of Haematology | 2016
Sophia Danhof; Martin Schreder; Leo Rasche; Susanne Strifler; Hermann Einsele; Stefan Knop
Carfilzomib, the second‐generation proteasome inhibitor, is still awaiting approval for the treatment of multiple myeloma in Europe. Results from clinical trials have demonstrated favorable efficacy in advanced disease but have opened an ongoing debate on cardiac complications related to carfilzomib treatment. ‘Real‐life’ data are scarce.
Blood | 2016
Niels Weinhold; Cody Ashby; Leo Rasche; Shweta S. Chavan; Caleb K. Stein; Owen Stephens; Ruslana Tytarenko; Michael Bauer; Tobias Meissner; Shayu Deshpande; Purvi Patel; Timea Buzder; Gabor Molnar; Erich Allen Peterson; van Rhee F; Maurizio Zangari; Sharmilan Thanendrarajan; Carolina Schinke; Erming Tian; Joshua Epstein; Bart Barlogie; Faith E. Davies; Christoph Heuck; Brian A. Walker; Gareth J. Morgan
To elucidate the mechanisms underlying relapse from chemotherapy in multiple myeloma, we performed a longitudinal study of 33 patients entered into Total Therapy protocols investigating them using gene expression profiling, high-resolution copy number arrays, and whole-exome sequencing. The study illustrates the mechanistic importance of acquired mutations in known myeloma driver genes and the critical nature of biallelic inactivation events affecting tumor suppressor genes, especially TP53, the end result being resistance to apoptosis and increased proliferation rates, which drive relapse by Darwinian-type clonal evolution. The number of copy number aberration changes and biallelic inactivation of tumor suppressor genes was increased in GEP70 high risk, consistent with genomic instability being a key feature of high risk. In conclusion, the study highlights the impact of acquired genetic events, which enhance the evolutionary fitness level of myeloma-propagating cells to survive multiagent chemotherapy and to result in relapse.
Nature Communications | 2017
Leo Rasche; Shweta S. Chavan; Owen Stephens; Purvi Patel; Ruslana Tytarenko; Cody Ashby; Michael Bauer; Caleb K. Stein; Shayu Deshpande; Christopher P. Wardell; Timea Buzder; Gabor Molnar; Maurizio Zangari; Fritz Van Rhee; Sharmilan Thanendrarajan; Carolina Schinke; Joshua Epstein; Faith E. Davies; Brian A. Walker; Tobias Meissner; Bart Barlogie; Gareth J. Morgan; Niels Weinhold
In multiple myeloma malignant plasma cells expand within the bone marrow. Since this site is well-perfused, a rapid dissemination of “fitter” clones may be anticipated. However, an imbalanced distribution of multiple myeloma is frequently observed in medical imaging. Here, we perform multi-region sequencing, including iliac crest and radiology-guided focal lesion specimens from 51 patients to gain insight into the spatial clonal architecture. We demonstrate spatial genomic heterogeneity in more than 75% of patients, including inactivation of CDKN2C and TP53, and mutations affecting mitogen-activated protein kinase genes. We show that the extent of spatial heterogeneity is positively associated with the size of biopsied focal lesions consistent with regional outgrowth of advanced clones. The results support a model for multiple myeloma progression with clonal sweeps in the early phase and regional evolution in advanced disease. We suggest that multi-region investigations are critical to understanding intra-patient heterogeneity and the evolutionary processes in multiple myeloma.In multiple myeloma, malignant cells expand within bone marrow. Here, the authors use multi-region sequencing in patient samples to analyse spatial clonal architecture and heterogeneity, providing novel insight into multiple myeloma progression and evolution.
Haematologica | 2015
Leo Rasche; Johannes Duell; Inês C. Castro; Valentina Dubljevic; Manik Chatterjee; Stefan Knop; Frank Hensel; Andreas Rosenwald; Hermann Einsele; Max S. Topp; Stephanie Brändlein
The primary objective of this phase 1 study was to evaluate the safety and tolerability of the anti-glucose regulated protein 78 monoclonal immunoglobulin M antibody PAT-SM6 in subjects with relapsed or refractory multiple myeloma. Twelve heavily pretreated patients received four intravenous infusions of PAT-SM6 at doses of 0.3, 1, 3, and 6 mg/kg within 2 weeks. Efficacy, pharmacokinetics and immunogenicity were followed up until the end of the trial (day 36). In addition, immune cell patterns in peripheral blood were assessed by flow cytometry and glucose regulated protein 78 expression status was evaluated in bone marrow specimens by immunohistochemistry and flow cytometry at screening. All doses administered were found to be safe and well tolerated; the maximum tolerated dose was not reached. The most common treatment emergent adverse event was leukopenia (grades 1 and 2) in eight out of the 12 multiple myeloma patients. Pharmacokinetic analysis demonstrated dose-proportional increases in drug serum concentration. The terminal half-life ranged from 5.86 to 8.41 h, the apparent volume of distribution ranged from 101 to 150 mL/kg, and clearance ranged from 8.11 to 16.1 mL/h/kg. All patients showed glucose regulated protein 78 surface expression on multiple myeloma cells. Four out of the 12 patients (33.3 %) had stable disease, according to the International Myeloma Working Group criteria, after PAT-SM6 treatment across the doses 1, 3 and 6 mg/kg. In summary, single-agent PAT-SM6 was well tolerated with modest clinical activity in relapsed or refractory multiple myeloma. Further trials exploring the combination of PAT-SM6 with existing myeloma therapies are planned. Trial registration: clinicaltrials.gov identifier: NCT01727778
British Journal of Haematology | 2015
Klaus Martin Kortüm; Christian Langer; Jorge Monge; Laura Bruins; Jan B. Egan; Yuan X. Zhu; Chang Xin Shi; Patrick Jedlowski; Jessica Schmidt; Juhi Ojha; Lars Bullinger; Peter Liebisch; Miriam Kull; Mia D. Champion; Scott Van Wier; Gregory J. Ahmann; Leo Rasche; Stefan Knop; Rafael Fonseca; Hermann Einsele; A. Keith Stewart; Esteban Braggio
We constructed a multiple myeloma (MM)‐specific gene panel for targeted sequencing and investigated 72 untreated high‐risk (del17p) MM patients. Mutations were identified in 78% of the patients. While the majority of studied genes were mutated at similar frequency to published literature, the prevalence of TP53 mutation was increased (28%) and no mutations were found in FAM46C. This study provides a comprehensive insight into the mutational landscape of del17p high‐risk MM. Additionally, our work demonstrates the practical use of a customized sequencing panel, as an easy, cheap and fast approach to characterize the mutational profile of MM.
Leukemia | 2018
Andrej Besse; S C Stolze; Leo Rasche; Niels Weinhold; Gareth J. Morgan; Marianne Kraus; J Bader; Herman S. Overkleeft; Lenka Besse; Christoph Driessen
Proteasome inhibitor (PI) carfilzomib (CFZ) has activity superior to bortezomib (BTZ) and is increasingly incorporated in multiple myeloma (MM) frontline therapy and relapsed settings. Most MM patients ultimately experience PI-refractory disease, an unmet medical need with poorly understood biology and dismal outcome. Pharmacologic targeting of ABCB1 improved patient outcomes, including MM, but suffered from adverse drug effects and insufficient plasma concentrations. Proteomics analysis identified ABCB1 overexpression as the most significant change in CFZ-resistant MM cells. We addressed the functional role of ABCB1 overexpression in MM and observed significantly upregulated ABCB1 in peripheral blood malignant plasma cells (PCs) vs untreated patients’ bone marrow PC. ABCB1 overexpression reduces the proteasome-inhibiting activity of CFZ due to drug efflux, in contrast to BTZ. Likewise, the cytotoxicity of established anti-MM drugs was significantly reduced in ABCB1-expressing MM cells. In search for potential drugs targeting ABCB1 in clinical trials, we identified the HIV protease inhibitors nelfinavir (NFV) and lopinavir (LPV) as potent functional modulators of ABCB1-mediated drug export, most likely via modulation of mitochondria permeability transition pore. NFV and LPV restored CFZ activity at therapeutically relevant drug levels and thus represent ready-to-use drugs to be tested in clinical trials to target ABCB1 and to re-sensitize PC to established myeloma drugs, in particular CFZ.
American Journal of Hematology | 2016
Artur Jurczyszyn; Norbert Grzasko; Alessandro Gozzetti; Jacek Czepiel; Alfonso Cerase; Vania Hungria; Edvan Crusoe; Ana Luiza Miranda Silva Dias; Ravi Vij; Mark Fiala; Jo Caers; Leo Rasche; Ajay K. Nooka; Sagar Lonial; David H. Vesole; Sandhya Philip; Shane Gangatharan; Agnieszka Druzd-Sitek; Jan Walewski; Alessandro Corso; Federica Cocito; Marie Christine M. Vekemans; Erden Atilla; Meral Beksac; Xavier Leleu; Julio Davila; Ashraf Badros; Ekta Aneja; Niels Abildgaard; Efstathios Kastritis
The multicenter retrospective study conducted in 38 centers from 20 countries including 172 adult patients with CNS MM aimed to describe the clinical and pathological characteristics and outcomes of patients with multiple myeloma (MM) involving the central nervous system (CNS). Univariate and multivariate analyses were performed to identify prognostic factors for survival. The median time from MM diagnosis to CNS MM diagnosis was 3 years. Thirty‐eight patients (22%) were diagnosed with CNS involvement at the time of initial MM diagnosis and 134 (78%) at relapse/progression. Upon diagnosis of CNS MM, 97% patients received initial therapy for CNS disease, of which 76% received systemic therapy, 36% radiotherapy and 32% intrathecal therapy. After a median follow‐up of 3.5 years, the median overall survival (OS) from the onset of CNS involvement for the entire group was 7 months. Untreated and treated patients had median OS of 2 and 8 months, respectively (P < 0.001). At least one previous line of therapy for MM before the diagnosis of CNS disease and >1 cytogenetic abnormality detected by FISH were independently associated with worse OS. The median OS for patients with 0, 1 and 2 of these risk factors were 25 months, 5.5 months and 2 months, respectively (P < 0.001). Neurological manifestations, not considered chemotherapy‐related, observed at any time after initial diagnosis of MM should raise a suspicion of CNS involvement. Although prognosis is generally poor, the survival of previously untreated patients and patients with favorable cytogenetic profile might be prolonged due to systemic treatment and/or radiotherapy. Am. J. Hematol. 91:575–580, 2016.
Blood Cancer Journal | 2017
Shweta S. Chavan; Jie He; Ruslana Tytarenko; Shayu Deshpande; Purvi Patel; Mark Bailey; Caleb K. Stein; Owen Stephens; Niels Weinhold; Nathan Petty; Douglas Steward; Leo Rasche; Michael Bauer; Cody Ashby; Erich Allen Peterson; Siraj M. Ali; Jeff Ross; Vincent A. Miller; P.J. Stephens; Sharmilan Thanendrarajan; Carolina Schinke; Maurizio Zangari; F van Rhee; B Barlogie; Tariq I. Mughal; Faith E. Davies; Gareth J. Morgan; Brian A. Walker
The purpose of this study is to identify prognostic markers and treatment targets using a clinically certified sequencing panel in multiple myeloma. We performed targeted sequencing of 578 individuals with plasma cell neoplasms using the FoundationOne Heme panel and identified clinically relevant abnormalities and novel prognostic markers. Mutational burden was associated with maf and proliferation gene expression groups, and a high-mutational burden was associated with a poor prognosis. We identified homozygous deletions that were present in multiple myeloma within key genes, including CDKN2C, RB1, TRAF3, BIRC3 and TP53, and that bi-allelic inactivation was significantly enriched at relapse. Alterations in CDKN2C, TP53, RB1 and the t(4;14) were associated with poor prognosis. Alterations in RB1 were predominantly homozygous deletions and were associated with relapse and a poor prognosis which was independent of other genetic markers, including t(4;14), after multivariate analysis. Bi-allelic inactivation of key tumor suppressor genes in myeloma was enriched at relapse, especially in RB1, CDKN2C and TP53 where they have prognostic significance.
Haematologica | 2017
Sharmilan Thanendrarajan; Erming Tian; Pingping Qu; Pankaj Mathur; Carolina Schinke; Frits van Rhee; Maurizio Zangari; Leo Rasche; Niels Weinhold; Daisy Alapat; William T. Bellamy; Cody Ashby; Sandra Mattox; Joshua Epstein; Shmuel Yaccoby; Bart Barlogie; Antje Hoering; Michael Bauer; Brian A. Walker; Faith E. Davies; Gareth J. Morgan
Multiple myeloma (MM) is a malignant disorder of plasma cells with a heterogeneous clinical outcome that is affected by both numerical and structural chromosomal abnormalities, baseline characteristics (age, lactate dehydrogenase concentration, International Staging System score) and treatment
Leukemia & Lymphoma | 2016
Artur Jurczyszyn; Magdalena Olszewska-Szopa; Vania Tietsche de Moraes Hungria; Edvan Crusoe; Tomas Pika; Michel Delforge; Xavier Leleu; Leo Rasche; Ajay K. Nooka; Agnieszka Druzd-Sitek; Jan Walewski; Julio Davila; Jo Caers; Vladimír Maisnar; Morie A. Gertz; Massimo Gentile; Dorotea Fantl; Giuseppe Mele; David H. Vesole; Andrew Yee; Chaim Shustik; Suzanne Lentzsch; Sonja Zweegman; Alessandro Gozzetti; Aleksander B. Skotnicki; Jorge J. Castillo
Abstract Skin infiltration in multiple myeloma (skin MM) is a rare clinical problem. Only a few cases of skin involvement have been reported, primarily in single case reports. We analyzed and present the clinical outcomes, immunohistochemistry and cytogenetic features, and relevant laboratory data on 53 biopsy-proven skin MM cases. The median time from MM diagnosis to skin involvement was 2 years. There appears to be an overrepresentation of immunoglobulin class A (IgA) and light chain disease in skin MM. We found no correlation between CD56 negative MM and skin infiltration. We found that skin MM patients presented in all MM stages (i.e. ISS stages I to III), and there was no preferential cytogenetic abnormality. Patients with skin MM carry a very poor prognosis with a median overall survival (OS) of 8.5 months as time from skin involvement. Moreover, patients with IgA disease and plasmablastic morphology appear to have a worse OS.