Network


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

Hotspot


Dive into the research topics where Christoph Heuck is active.

Publication


Featured researches published by Christoph Heuck.


Journal of Hematology & Oncology | 2009

Mechanism of action of lenalidomide in hematological malignancies

Venumadhav Kotla; Swati Goel; Sangeeta Nischal; Christoph Heuck; Kumar Vivek; Bhaskar C. Das; Amit Verma

Immunomodulatory drugs lenalidomide and pomalidomide are synthetic compounds derived by modifying the chemical structure of thalidomide to improve its potency and reduce its side effects. Lenalidomide is a 4-amino-glutamyl analogue of thalidomide that lacks the neurologic side effects of sedation and neuropathy and has emerged as a drug with activity against various hematological and solid malignancies. It is approved by FDA for clinical use in myelodysplastic syndromes with deletion of chromosome 5q and multiple myeloma. Lenalidomide has been shown to be an immunomodulator, affecting both cellular and humoral limbs of the immune system. It has also been shown to have anti-angiogenic properties. Newer studies demonstrate its effects on signal transduction that can partly explain its selective efficacy in subsets of MDS. Even though the exact molecular targets of lenalidomide are not well known, its activity across a spectrum of neoplastic conditions highlights the possibility of multiple target sites of action.


Blood | 2012

Stem and progenitor cells in myelodysplastic syndromes show aberrant stage-specific expansion and harbor genetic and epigenetic alterations

Britta Will; Li Zhou; Thomas O. Vogler; Susanna Ben-Neriah; Carolina Schinke; Roni Tamari; Yiting Yu; Tushar D. Bhagat; Sanchari Bhattacharyya; Laura Barreyro; Christoph Heuck; Yonkai Mo; Samir Parekh; Christine McMahon; Andrea Pellagatti; Jacqueline Boultwood; Cristina Montagna; Lewis B. Silverman; Jaroslaw P. Maciejewski; John M. Greally; B. Hilda Ye; Alan F. List; Christian Steidl; Ulrich Steidl; Amit Verma

Even though hematopoietic stem cell (HSC) dysfunction is presumed in myelodysplastic syndrome (MDS), the exact nature of quantitative and qualitative alterations is unknown. We conducted a study of phenotypic and molecular alterations in highly fractionated stem and progenitor populations in a variety of MDS subtypes. We observed an expansion of the phenotypically primitive long-term HSCs (lineage(-)/CD34(+)/CD38(-)/CD90(+)) in MDS, which was most pronounced in higher-risk cases. These MDS HSCs demonstrated dysplastic clonogenic activity. Examination of progenitors revealed that lower-risk MDS is characterized by expansion of phenotypic common myeloid progenitors, whereas higher-risk cases revealed expansion of granulocyte-monocyte progenitors. Genome-wide analysis of sorted MDS HSCs revealed widespread methylomic and transcriptomic alterations. STAT3 was an aberrantly hypomethylated and overexpressed target that was validated in an independent cohort and found to be functionally relevant in MDS HSCs. FISH analysis demonstrated that a very high percentage of MDS HSC (92% ± 4%) carry cytogenetic abnormalities. Longitudinal analysis in a patient treated with 5-azacytidine revealed that karyotypically abnormal HSCs persist even during complete morphologic remission and that expansion of clonotypic HSCs precedes clinical relapse. This study demonstrates that stem and progenitor cells in MDS are characterized by stage-specific expansions and contain epigenetic and genetic alterations.


Cancer Research | 2011

Reduced SMAD7 Leads to Overactivation of TGF-β Signaling in MDS that Can Be Reversed by a Specific Inhibitor of TGF-β Receptor I Kinase

Li Zhou; Christine McMahon; Tushar D. Bhagat; Cristina Alencar; Yiting Yu; Melissa Fazzari; Davendra Sohal; Christoph Heuck; Krishna Gundabolu; Chun Ng; Yongkai Mo; Wa Shen; Amittha Wickrema; Guanghui Kong; Ellen Friedman; Lubomir Sokol; Giannis Mantzaris; Andrea Pellagatti; Jacqueline Boultwood; Leonidas C. Platanias; Ulrich Steidl; Lei Yan; Jonathan M. Yingling; Michael Lahn; Alan F. List; Markus Bitzer; Amit Verma

Even though myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis, the molecular alterations that lead to marrow failure have not been well elucidated. We have previously shown that the myelosuppressive TGF-β pathway is constitutively activated in MDS progenitors. Because there is conflicting data about upregulation of extracellular TGF-β levels in MDS, we wanted to determine the molecular basis of TGF-β pathway overactivation and consequent hematopoietic suppression in this disease. We observed that SMAD7, a negative regulator of TGF-β receptor I (TBRI) kinase, is markedly decreased in a large meta-analysis of gene expression studies from MDS marrow-derived CD34(+) cells. SMAD7 protein was also found to be significantly decreased in MDS marrow progenitors when examined immunohistochemically in a bone marrow tissue microarray. Reduced expression of SMAD7 in hematopoietic cells led to increased TGF-β-mediated gene transcription and enhanced sensitivity to TGF-β-mediated suppressive effects. The increased TGF-β signaling due to SMAD7 reduction could be effectively inhibited by a novel clinically relevant TBRI (ALK5 kinase) inhibitor, LY-2157299. LY-2157299 could inhibit TGF-β-mediated SMAD2 activation and hematopoietic suppression in primary hematopoietic stem cells. Furthermore, in vivo administration of LY-2157299 ameliorated anemia in a TGF-β overexpressing transgenic mouse model of bone marrow failure. Most importantly, treatment with LY-2157199 stimulated hematopoiesis from primary MDS bone marrow specimens. These studies demonstrate that reduction in SMAD7 is a novel molecular alteration in MDS that leads to ineffective hematopoiesis by activating of TGF-β signaling in hematopoietic cells. These studies also illustrate the therapeutic potential of TBRI inhibitors in MDS.


Blood | 2012

Second malignancies in total therapy 2 and 3 for newly diagnosed multiple myeloma: influence of thalidomide and lenalidomide during maintenance

Saad Z Usmani; Rachel Sexton; Antje Hoering; Christoph Heuck; Bijay Nair; Sarah Waheed; Yazan Al Sayed; Nabeel Chauhan; Nisar Ahmad; Shebli Atrash; Nathan Petty; Frits van Rhee; John Crowley; Bart Barlogie

Thalidomide and lenalidomide constitute an important part of effective myeloma therapy. Recent data from the Intergroup Francophone du Myélome, Cancer and Leukemia Group B, and Gruppo Italiano Malattie Ematologiche dell Adulto MM-015 trials suggest that lenalidomide maintenance therapy is associated with a higher incidence of second primary malignancies (SPMs), including both hematologic and solid malignancies. In the present study, we analyzed data from the Total Therapy 2 (TT2) trial, along with the 2 Total Therapy 3 (TT3) trials. TT2 patients were assigned randomly to either a control group (no thalidomide) or to the experimental group (thalidomide during induction, between transplantations, and during consolidation and maintenance). The 2 TT3 trials used thalidomide and bortezomib during induction, before and in consolidation after tandem melphalan-based transplantation; TT3A applied VTD (bortezomib, thalidomide, dexamethasone) in the first year of maintenance and TD for 2 more years, whereas TT3B used VRD (bortezomib, lenalidomide, dexamethasone) maintenance for 3 years. The cumulative incidence of SPMs did not differ significantly among the TT trial components when measured from enrollment (P = .78) or from initiation of maintenance (P = .82). However, a pairwise comparison of the TT2 arms suggested a lower incidence of hematologic SPMs in the thalidomide maintenance arm (hazard ratio = 0.38; P = .09). These trials are registered at www.clinicaltrials.gov as NCT00573391 (TT2), NCT00081939 (TT3A), and NCT00572169 (TT3B).


Journal of Immunology | 2013

Myeloma Is Characterized by Stage-Specific Alterations in DNA Methylation That Occur Early during Myelomagenesis

Christoph Heuck; Jayesh Mehta; Tushar D. Bhagat; Krishna Gundabolu; Yiting Yu; Shahper N. Khan; Grigoris Chrysofakis; Carolina Schinke; Joseph Tariman; Eric Vickrey; Natalie Pulliam; Sangeeta Nischal; Li Zhou; Sanchari Bhattacharyya; Richard Meagher; Caroline Hu; Shahina Maqbool; Masako Suzuki; Samir Parekh; Frederic J. Reu; Ulrich Steidl; John M. Greally; Amit Verma; Seema Singhal

Epigenetic changes play important roles in carcinogenesis and influence initial steps in neoplastic transformation by altering genome stability and regulating gene expression. To characterize epigenomic changes during the transformation of normal plasma cells to myeloma, we modified the HpaII tiny fragment enrichment by ligation–mediated PCR assay to work with small numbers of purified primary marrow plasma cells. The nano-HpaII tiny fragment enrichment by ligation–mediated PCR assay was used to analyze the methylome of CD138+ cells from 56 subjects representing premalignant (monoclonal gammopathy of uncertain significance), early, and advanced stages of myeloma, as well as healthy controls. Plasma cells from premalignant and early stages of myeloma were characterized by striking, widespread hypomethylation. Gene-specific hypermethylation was seen to occur in the advanced stages, and cell lines representative of relapsed cases were found to be sensitive to decitabine. Aberrant demethylation in monoclonal gammopathy of uncertain significance occurred primarily in CpG islands, whereas differentially methylated loci in cases of myeloma occurred predominantly outside of CpG islands and affected distinct sets of gene pathways, demonstrating qualitative epigenetic differences between premalignant and malignant stages. Examination of the methylation machinery revealed that the methyltransferase, DNMT3A, was aberrantly hypermethylated and underexpressed, but not mutated in myeloma. DNMT3A underexpression was also associated with adverse overall survival in a large cohort of patients, providing insights into genesis of hypomethylation in myeloma. These results demonstrate widespread, stage-specific epigenetic changes during myelomagenesis and suggest that early demethylation can be a potential contributor to genome instability seen in myeloma. We also identify DNMT3A expression as a novel prognostic biomarker and suggest that relapsed cases can be therapeutically targeted by hypomethylating agents.


Wiley Interdisciplinary Reviews: Systems Biology and Medicine | 2010

DNA methylation alterations in multiple myeloma as a model for epigenetic changes in cancer

Amy Sharma; Christoph Heuck; Melissa Fazzari; Jayesh Mehta; Seema Singhal; John M. Greally; Amit Verma

Epigenetics refers to heritable modifications of the genome that are not a result of changes in the DNA sequence and result in phenotypic changes. These changes can be stably transmitted through cell division and are potentially reversible. Epigenetic events are very important during normal development wherein a single progenitor cell proliferates and differentiates into various somatic cell types. This process occurs through modification of the genome without changing the genetic code. Because epigenetic control of gene expression is so important, aberrant epigenetic regulation can lead to disease and cancer. This article reviews epigenetic changes seen in cancer by examining epigenetic changes commonly found in multiple myeloma, a common hematologic malignancy of plasma cells. Epigenetic control of gene expression can be exerted by changes in DNA methylation, histone modifications, and expression of noncoding RNAs. Each of these regulatory mechanisms interacts with the others at different genomic locations and can be measured quantitatively within the cell, requiring that we consider these mechanisms not individually but as a biological system. DNA methylation was the earliest discovered epigenetic regulator and has been the focus of most investigations in cancer. We have thus focused on DNA methylation changes in the pathogenesis of multiple myeloma, which promises to become an excellent model for systems biological studies of epigenomic dysregulation in human disease. Copyright


Leukemia | 2012

Primary plasma cell leukemia: clinical and laboratory presentation, gene-expression profiling, and clinical outcome with Total Therapy protocols

Saad Z Usmani; Bijay Nair; Pingping Qu; Emily Hansen; Qing Zhang; Nathan Petty; Sarah Waheed; John D. Shaughnessy; Yazan Alsayed; Christoph Heuck; Frits van Rhee; Teresa Milner; Antje Hoering; Jackie Szymonifka; Rachael Sexton; Jeffrey R. Sawyer; Zeba N. Singh; John Crowley; Bart Barlogie

To determine whether primary plasma cell leukemia (PPCL) remains a high-risk multiple myeloma feature in the context of contemporary therapy and gene-expression profiling (GEP), we reviewed records of 1474 patients with myeloma, who were enrolled in Total Therapy protocols or treated identically off protocol. A total of 27 patients (1.8%) were classified as having PPCL. As a group, these patients more often had low hemoglobin, high beta-2-microglobulin, high lactate dehydrogenase, low albumin and cytogenetic abnormalities. Among 866 patients with GEP results, the PPCL group more often had disease that was classified as high risk, and in CD-1 and MF molecular subgroups. Regardless of the therapeutic protocol, patients with PPCL had shorter median overall survival (OS; 1.8 years), progression-free survival (PFS; 0.8 years) and complete response duration (CRD; 1.3 years) than the remainder, whose clinical outcomes had improved markedly with successive protocols. Multivariate analyses of pretreatment parameters showed that PPCL was a highly significant independent adverse feature linked to OS, PFS and CRD. In GEP analyses, 203 gene probes distinguished PPCL from non-PPCL; the identified genes were involved in the LXR/RXR activation, inositol metabolism, hepatic fibrosis/hepatic stellate-cell activation and lipopolysaccharide/interleukin-1-mediated inhibition of RXR function pathways. Different treatment approaches building on these genomic differences may improve the grave outcome of patients with PPCL.


Blood | 2016

Clonal selection and double-hit events involving tumor suppressor genes underlie relapse in myeloma.

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.


PLOS ONE | 2013

Characterization of the molecular mechanism of the bone-anabolic activity of carfilzomib in multiple myeloma.

Bo Hu; Yu Chen; Saad Z Usmani; Shiqiao Ye; Wei Qiang; Xenofon Papanikolaou; Christoph Heuck; Shmuel Yaccoby; Bart O. Williams; Frits van Rhee; Bart Barlogie; Joshua Epstein; Ya-Wei Qiang

Carfilzomib, the next generation of proteasome inhibitor, may increase osteoblast-related markers in patients with multiple myeloma, but the molecular mechanism of its effect on mesenchymal stem cell differentiation to osteoblasts remains unknown. Herein, we demonstrated that carfilzomib significantly promoted mesenchymal stem cell differentiation into osteoblasts. In osteoprogenitor cells and primary mesenchymal stem cells from patients with myeloma, carfilzomib induced increases in alkaline phosphatase activity, matrix mineralization, and calcium deposition via Wnt-independent activation of β-catenin/TCF signaling. Using affinity pull-down assays with immunoblotting analysis and immunofluorescence, we found that carfilzomib induced stabilization of both free and active forms of β-catenin in a time- and dose-dependent manner that was not associated with β-catenin transcriptional regulation. Nuclear translocation of β-catenin protein was associated with TCF transcriptional activity that was independent of the effects of GSK3β-activation and of signaling induced by 19 Wnt ligands, 10 Frizzled receptors, and LRP5/6 co-receptors. Blocking activation of β-catenin/TCF signaling by dominant negative TCF1 or TCF4 attenuated carfilzomib-induced matrix mineralization. Thus, carfilzomib induced osteoblast differentiation via Wnt-independent activation of the β-catenin/TCF pathway. These results provide a novel molecular mechanism critical to understanding the anabolic role of carfilzomib on myeloma-induced bone disease.


International Journal of Hematology | 2011

The use of molecular-based risk stratification and pharmacogenomics for outcome prediction and personalized therapeutic management of multiple myeloma

Sarah K. Johnson; Christoph Heuck; Anthony P. Albino; Pingping Qu; Qing Zhang; Bart Barlogie; John D. Shaughnessy

Despite improvement in therapeutic efficacy, multiple myeloma (MM) remains incurable with a median survival of approximately 10 years. Gene-expression profiling (GEP) can be used to elucidate the molecular basis for resistance to chemotherapy through global assessment of molecular alterations that exist at diagnosis, after therapeutic treatment and that evolve during tumor progression. Unique GEP signatures associated with recurrent chromosomal translocations and ploidy changes have defined molecular classes with differing clinical features and outcomes. When compared to other stratification systems the GEP70 test remained a significant predictor of outcome, reduced the number of patients classified with a poor prognosis, and identified patients at increased risk of relapse despite their standard clinico-pathologic and genetic findings. GEP studies of serial samples showed that risk increases over time, with relapsed disease showing GEP shifts toward a signature of poor outcomes. GEP signatures of myeloma cells after therapy were prognostic for event-free and overall survival and thus may be used to identify novel strategies for overcoming drug resistance. This brief review will focus on the use of GEP of MM to define high-risk myeloma, and elucidate underlying mechanisms that are beginning to change clinical decision-making and inform drug design.

Collaboration


Dive into the Christoph Heuck's collaboration.

Top Co-Authors

Avatar

Bart Barlogie

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Frits van Rhee

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Joshua Epstein

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Gareth J. Morgan

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Maurizio Zangari

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Sarah Waheed

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Faith E. Davies

University of Arkansas for Medical Sciences

View shared research outputs
Top Co-Authors

Avatar

Antje Hoering

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Saad Z Usmani

Carolinas Healthcare System

View shared research outputs
Top Co-Authors

Avatar

Donald Johann

University of Arkansas for Medical Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge