• About
CMM
Karolinska institutet
Karolinska universitetssjukhuset
Sök

Richard Rosenquist Brandell Group

Clinical Genetics

About

We investigate genetic disorders on a molecular level in a translational research setting in order to understand disease mechanisms, apply new knowledge in genetic diagnostics, improve prognosis assessment and develop new treatment strategies. Our research group consists of four teams and our main interests are hematological malignancies, with a particular focus on chronic lymphocytic leukemia, lymphoma and pediatric acute lymphoblastic leukemia, metabolic bone disorders, and cancer genetics.

Research areas

Chronic lymphocytic leukemia (CLL) / Lymphoma

PI: Professor Richard Rosenquist Brandell

Chronic lymphocytic leukemia (CLL) is characterized by accumulation of monoclonal B cells in secondary lymphoid organs, bone marrow and peripheral blood. The median age at diagnosis is 71 years and men are more frequently affected than women. The disease is clinically and biologically heterogeneous, ranging from indolent with no treatment requirement, to a very aggressive disease characterized by chemorefractoriness and poor survival. In clinical practice, two staging systems are used (Rai and Binet) however, despite their clinical utility, they are unable to predict which patients with low tumor burden will experience an aggressive disease as opposed to an indolent course. There are currently several molecular markers that stratify CLL into prognostic subgroups, the most important of which are the mutational status of the immunoglobulin heavy variable (IGHV) genes, as well as recurrent chromosomal aberrations, where deletion of 17p (harboring the TP53 gene) is associated with an aggressive disease course and poor outcome. To this day CLL remains incurable but novel therapies using antibodies and small molecule inhibitors have advanced the field substantially.

Four main research areas

Our research projects can be subdivided in four main areas. These are:

1. Defining the molecular make-up of CLL subsets expressing stereotyped B-cell receptors, which currently represent the most meaningful homogeneous subgroups to investigate. We use multiple high throughput, next-generation sequencing (NGS) technologies, including whole-genome-, transcriptome- and single-cell sequencing, as well as high-resolution methylation arrays in a clinically well-characterized CLL cohort that express stereotyped B-cell receptors. Functional analyses are performed on selected mutations/transcripts to study their effects at a cellular level by utilizing knock-down (shRNA, CRISPR/Cas9) or transfection assays in CLL cell lines.

2. Identifying dysregulated intracellular signaling pathways and processes in distinct CLL subsets. We use different OMICs approaches including RNA-sequencing and proteomics that enables in-depth analyses of dysregulated signaling pathways in CLL. We also apply phospho-flow cytometry that measures protein phosphorylation events at single cell level and allows for simultaneous analysis of multiple signaling proteins and pathways. Focus is on B-cell receptor, MAPK/ERK, STAT and NF-kB signaling pathways.

3. Understanding resistance mechanisms in CLL using NGS and high throughput drug screening, with a focus on patients that relapse within a short period of receiving therapy. We combine whole-exome/genome sequencing in primary cells in pre- and post-treated samples from patients receiving small molecule inhibitors. Additionally, we carry out comprehensive high throughput drug testing on tumor cells from CLL patients relapsing after receiving targeted therapy in order to systematically identify drug sensitivities and resistance patterns.

4. Validating, harmonizing and implementing NGS-based assays for clinical routine diagnostics. For this purpose, we work together with the European Research Initiative in CLL (ERIC) and perform multicenter validation of novel genetic findings and assays. We assess the clinical impact of recurrent mutations in large well-annotated international series of CLL cases for which both molecular and clinical characteristics are available. Once validated, new biomarkers or assays can be transferred at a national level to routine diagnostics, using the SciLifeLab Diagnostics Development platform or Genomic Medicine Sweden.

Pediatric leukemia

PI: Associate Professor Gisela Barbany

The focus of the pediatric leukemia team is translational research in malignant hematology, in particular genetic characterization of pediatric acute lymphoblastic leukemia (ALL). The overall goal of the group is to improve the tools used in the stratification of ALL patients. For that purpose, we are working with four different subprojects:

1. Implement next-generation sequencing techniques in the diagnostic setting of ALL and validate whole-genome sequencing (WGS) as a routine diagnostic procedure for acute leukemia. We also develop quantitative methods using the unique sequences generated by structural events in the leukemic blasts to monitor therapy response.

2. We investigate particular genetic subgroups of ALL to understand the impact on blast biology and behavior of the driving genetic event.

3. We study ALL patients lacking stratifying genetic markers and try to identify and validate novel potential genetic markers.

4. We investigate the pathogenic mechanisms underlying the genetic events that recurrently affect particular regions of the genome in acute leukemia.

Significance: Through the combination of different high-throughput methodologies, our studies will contribute to understand the consequences of genetic aberrations on leukemia biology and behavior as well as the mechanisms that make these chromosome regions prone to recurrently engage in complex rearrangements. We expect that our studies combined will improve the diagnostic procedures that underlie risk stratification of ALL patients and thus contribute to decrease over- and undertreatment of children with ALL.

Metabolic bone diseases

PI: Professor Outi Mäkitie

Our team studies genetic defects and molecular mechanisms underlying skeletal disorders, with main focus on early-onset primary osteoporosis, skeletal dysplasias, and vitamin D.

Osteoporosis is a skeletal disorder characterized by reduced bone mineral density, compromised bone strength, and susceptibility to fractures. Genetic factors play a major role in disease susceptibility and characteristics. Due to the silent development of the disease, diagnosis is usually made at adult age when the severe consequences of bone fragility emerge, but the disease often has its onset in childhood. The pathomolecular mechanisms leading to disease in young patients with novel forms of osteoporosis have yet to be fully explored. Moreover, evidence on efficacy of currently available treatments for osteoporosis in children and young adults is still lacking.

Skeletal dysplasias encompass a group of more than 400 monogenic diseases with significant skeletal involvement. Due to the rarity of these conditions and their broad clinical and genetic heterogeneity, many families still lack a genetic diagnosis and novel gene defects remain to be characterized. The lack of pharmacological treatment options for most subtypes of skeletal dysplasia is a further challenge and merits better understanding of disease mechanisms.

Vitamin D deficiency is a significant health concern in several populations. Recent studies by others and us have shown that especially children with a chronic illness, including childhood cancer, are at high risk of vitamin D deficiency. This in turn may have significant health consequences, including impaired skeletal health.

By combining genetic testing, primarily next-generation sequencing, and in vitro functional studies our aims are:

1. To characterize novel variants and genes involved in skeletal development and maintenance. We have collected detailed clinical information and genomic DNA from several families affected by early-onset osteoporosis and some subtypes of skeletal dysplasias with unknown genetic causes. By applying whole-genome sequencing, we aim to pinpoint the genetic cause of disease in these families. Once a novel candidate variant is identified, molecular studies at RNA and protein levels are carried out to elucidate the biological effects of the identified gene variants.

2. To decipher the role of novel and previously identified gene defects. Cell biological disease mechanisms are investigated by studying patient-derived cells. Skin-derived fibroblasts from several families with rare skeletal diseases as well as bone marrow from a handful of patients have been collected. Different in vitro assays are carried out to study the pathomechanisms based on specific research questions and function of the investigated gene. Part of our research focus on skeletal progenitors (mesenchymal stromal cells, MSCs) and their role in skeletal cell differentiation and function in metabolic bone diseases.

3. To elucidate the role of Vitamin D in bone health. We have an ongoing large-scale vitamin D intervention study in infants (VIDI) that aims to find means to optimize vitamin D status in young children. Genetic data from this cohort allow us to perform genome-wide association studies to search for novel genetic loci associated with circulating vitamin D levels and bone health in a prospective longitudinal setting.

Significance: By identifying novel disease-gene associations and by understanding the disease pathogenesis we will establish means for early diagnosis, and better prevention and treatment of childhood-onset skeletal disorders.

Cancer genetics

PI: Emma Tham, Senior researcher

Genetic alterations underlie all forms of cancer: in most cases, these changes are somatic and give rise to sporadic cancer. In 10% of cases, they are inherited and constitutional and cause hereditary cancer. Individuals carrying a predisposing genetic variant have an increased risk of developing cancer (and sometimes other symptoms) and benefit from prevention schemes. However, as most hereditary cancer is rare, knowledge regarding the causative genes, as well as evidence regarding cancer risks and surveillance are lacking.

When tumor cells die, they release their DNA into body fluids such as blood and thus it is possible to detect their genetic alterations in a blood sample (so called liquid biopsy). These cell-free tumor DNA (ctDNA) fragments make up a minor proportion of the cell free DNA fragments that mostly derive from the blood cells and therefore, very sensitive methods are required in order to detect them. ctDNA has the potential to revolutionize cancer diagnostics and are of particular relevance in high-risk individuals with cancer predisposition. ctDNA may also serve as a predictive biomarker that can be used to monitor therapy response and minimal residual disease in sporadic cancers.

Our research involves the following studies:

The Cancer Predisposition study (CAP) aims to systematically include all individuals with rare hereditary cancer, with and without a genetic cause. The aims are:

1. To discover new genetic causes for hereditary cancer using the novel massive parallel DNA/RNA sequencing techniques

2. To characterize rare cancer syndromes in order to offer personalized prevention

SWEP53: Molecular Characterization and Clinical Aspects of Germline TP53 Mutations in the Swedish Constitutional TP53 Cohort (national study, ISRCTN13103571). Collaboration project with Associate Prof. Svetlana Lagercrantz, Dept of Oncology and Pathology. The aims are to:

1. Characterize the constitutional TP53 cohort in Sweden

2. Evaluate the medical and psychosocial effects of rapid whole-body; brain and breast MRI as surveillance

3. Evaluate liquid biopsy as a complement to clinical and radiological surveillance

Cell-free tumor DNA for early diagnosis, prognosis and monitoring of cancer. The aims are to:

1. Evaluate ctDNA as a complement to clinical and radiological surveillance in individuals with hereditary predisposition for cancer

2. Evaluate ctDNA as a prognostic biomarker prior to surgery in patients with cancer

3. Evaluate ctDNA as a complement to clinical and radiological surveillance in following therapy response, monitoring minimal residual disease and detecting relapse       

Our team also participates in other studies that aim to improve the diagnosis of hereditary cancer through optimized genetic characterization of specific cancer types and improved genetic counselling.

We work closely with Clinical Genetics at Karolinska University Hospital on molecular diagnosis and monitoring of hematological malignancies and investigation of patients with hereditary cancer. We are also active in Genomic Medicine Sweden (GMS) and Genomic Medicine Centre Karolinska (GMCK) within solid tumors and liquid biopsy and involved in the European Reference Network for Genetic Tumor Risk Syndromes (GENTURIS).

Group Leader

Richard Rosenquist Brandell was appointed Professor of Clinical Genetics at the Department of Molecular Medicine and Surgery, Karolinska Institutet and Senior Physician in Clinical Genetics at Karolinska University Hospital, Sweden in 2017. He received his medical degree (1996) and PhD degree (1998) at Umeå University, Sweden, undertook a postdoctoral period at the Department of Pathology, Frankfurt am Main, Germany, and became specialist in Clinical Genetics 2004. Richard Rosenquist Brandell started his own research group at Uppsala University in 2000, focusing on molecular characterization of lymphoid malignancies, and his group rapidly became internationally renowned. He became Professor of Molecular Hematology in 2007 at the Department of Immunology, Genetics and Pathology, Uppsala University. He has initiated and led the SciLifeLab Clinical Genomics Facility in Uppsala between 2013-2017 and is currently Platform Director for the national Diagnostics Development Platform within SciLifeLab. More recently, he is coordinating the Genomic Medicine Sweden initiative that aims to build a new type of infrastructure within Swedish healthcare that implements precision medicine at a national level.

By employing a translational approach and utilizing cutting‐edge molecular tools, including next-generation sequencing technologies, Richard Rosenquist Brandell has made outstanding contributions to our understanding of the mechanisms behind the development of chronic lymphocytic leukemia (CLL), the most common adult leukemia. His studies have identified novel prognostic and predictive markers, defined new clinically relevant CLL subgroups, as well as provided significantly improved risk stratification at the individual patient level. Even more importantly, Rosenquist Brandell’s team has presented compelling evidence for a role of antigens (both autoantigens and microbial antigens) in the pathogenesis of CLL, which has generated great interest internationally. He has also built competitive networks at the national, European and international level. He is one of five founding members of an eminent European network of CLL researcher with an impressive combined cohort of more than 34,000 CLL patients from 24 academic institutions.

Richard Rosenquist Brandell has successfully supervised 23 PhD students as well as seven postdocs, and his research has resulted in more than 200 peer-reviewed papers. He was recently selected as Wallenberg Clinical Scholars 2017 by the Knut and Alice Wallenberg Foundation. 

Group members

Genomic Medicine Sweden

Richard Rosenquist Brandell is the director of Genomic Medicine Sweden (GMS) initiative that aims to build a new type of infrastructure within Swedish healthcare that implements precision medicine at a national level. This is the link to the GMS website.

Selected publications

See the full list of publications here.

About CMM

The Center for Molecular Medicine (CMM) is a foundation instituted by the Stockholm County Council (Region Stockholm). CMM is at the heart of a close partnership with the Karolinska University Hospital and Karolinska Institutet, fueling advancements in biomedical and clinical research.

Contact

Center for Molecular Medicine Foundation, org. nr. 815201-3689

Karolinska University Hospital L8:05

Visionsgatan 18

171 76 Stockholm, Sweden

communication@cmm.se

CMM
Karolinska institutet
Karolinska universitetssjukhuset