Network


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

Hotspot


Dive into the research topics where Mikael L. Rinne is active.

Publication


Featured researches published by Mikael L. Rinne.


Nucleic Acids Research | 2005

N-methylpurine DNA glycosylase overexpression increases alkylation sensitivity by rapidly removing non-toxic 7-methylguanine adducts

Mikael L. Rinne; Ying-Hui He; Brian F. Pachkowski; Jun Nakamura; Mark R. Kelley

Previous studies indicate that overexpression of N-methylpurine DNA glycosylase (MPG) dramatically sensitizes cells to alkylating agent-induced cytotoxicity. We recently demonstrated that this sensitivity is preceded by an increased production of AP sites and strand breaks, confirming that overexpression of MPG disrupts normal base excision repair and causes cell death through overproduction of toxic repair intermediates. Here we establish through site-directed mutagenesis that MPG-induced sensitivity to alkylation is dependent on enzyme glycosylase activity. However, in contrast to the sensitivity seen to heterogeneous alkylating agents, MPG overexpression generates no cellular sensitivity to MeOSO2(CH2)2-lexitropsin, an alkylator which exclusively induces 3-meA lesions. Indeed, MPG overexpression has been shown to increase the toxicity of alkylating agents that produce 7-meG adducts, and here we demonstrate that MPG-overexpressing cells have dramatically increased removal of 7-meG from their DNA. These data suggest that the mechanism of MPG-induced cytotoxicity involves the conversion of non-toxic 7-meG lesions into highly toxic repair intermediates. This study establishes a mechanism by which a benign DNA modification can be made toxic through the overexpression of an otherwise well-tolerated gene product, and the application of this principle could lead to improved chemotherapeutic strategies that reduce the peripheral toxicity of alkylating agents.


The journal of supportive oncology | 2012

Central Nervous System Complications of Cancer Therapy

Mikael L. Rinne; Eudocia Q. Lee; Patrick Y. Wen

As more effective therapies prolong the survival of patients with cancer, therapy-related toxicities, particularly those affecting the central nervous system (CNS) become increasingly important. CNS complications can cause significant morbidity and can limit the dose or duration of otherwise effective treatments. Because effects on the CNS are disabling and often permanent and treatments remain limited, it is important that clinicians recognize the effects of cancer therapy on the CNS. Cytotoxic chemotherapy and radiation are well-known causes of neurotoxicity, but there is increasing recognition that novel therapies are also sources of adverse effects on the CNS. This review highlights the CNS complications that result from radiation, chemotherapy, and novel therapeutics.


Neuro-oncology | 2015

Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma

Eudocia Q. Lee; David A. Reardon; David Schiff; Jan Drappatz; Alona Muzikansky; Sean Grimm; Andrew D. Norden; Lakshmi Nayak; Rameen Beroukhim; Mikael L. Rinne; Andrew S. Chi; Tracy T. Batchelor; Kelly Hempfling; Christine McCluskey; Katrina H. Smith; Sarah C. Gaffey; Brendan Wrigley; Keith L. Ligon; Jeffrey Raizer; Patrick Y. Wen

BACKGROUND Panobinostat is a histone deacetylase inhibitor with antineoplastic and antiangiogenic effects in glioma that may work synergistically with bevacizumab. We conducted a multicenter phase II trial of panobinostat combined with bevacizumab in patients with recurrent high-grade glioma (HGG). METHODS Patients with recurrent HGG were treated with oral panobinostat 30 mg 3 times per week, every other week, in combination with bevacizumab 10 mg/kg every other week. The primary endpoint was a 6-month progression-fee survival (PFS6) rate for participants with recurrent glioblastoma (GBM). Patients with recurrent anaplastic glioma (AG) were evaluated as an exploratory arm of the study. RESULTS At interim analysis, the GBM arm did not meet criteria for continued accrual, and the GBM arm was closed. A total of 24 patients with GBM were accrued prior to closure. The PFS6 rate was 30.4% (95%, CI 12.4%-50.7%), median PFS was 5 months (range, 3-9 months), and median overall survival (OS) was 9 months (range, 6-19 months). Accrual in the AG arm continued to completion, and a total of 15 patients were enrolled. The PFS6 rate was 46.7% (range, 21%-73%), median PFS was 7 months (range, 2-10 months), and median OS was 17 months (range, 5 months-27 months). CONCLUSIONS This phase II study of panobinostat and bevacizumab in participants with recurrent GBM did not meet criteria for continued accrual, and the GBM cohort of the study was closed. Although it was reasonably well tolerated, the addition of panobinostat to bevacizumab did not significantly improve PFS6 compared with historical controls of bevacizumab monotherapy in either cohort.


Cancer | 2015

Extent of resection and overall survival for patients with atypical and malignant meningioma

Ayal A. Aizer; Wenya Linda Bi; Manjinder S. Kandola; Eudocia Q. Lee; Lakshmi Nayak; Mikael L. Rinne; Andrew D. Norden; Rameen Beroukhim; David A. Reardon; Patrick Y. Wen; Ossama Al-Mefty; Nils D. Arvold; Ian F. Dunn; Brian M. Alexander

The prognosis for patients with atypical and malignant meningioma is guarded; whether the extent of resection is associated with survival‐based outcomes in this population remains poorly defined. This study investigated the association between gross total resection (GTR) and all‐cause mortality in patients with atypical and malignant meningioma.


Neuro-oncology | 2014

Adjuvant radiation therapy, local recurrence, and the need for salvage therapy in atypical meningioma

Ayal A. Aizer; Nils D. Arvold; Paul J. Catalano; Elizabeth B. Claus; Alexandra J. Golby; Mark D. Johnson; Ossama Al-Mefty; Patrick Y. Wen; David A. Reardon; Eudocia Q. Lee; Lakshmi Nayak; Mikael L. Rinne; Rameen Beroukhim; Stephanie E. Weiss; Shakti Ramkissoon; Malak Abedalthagafi; Sandro Santagata; Ian F. Dunn; Brian M. Alexander

BACKGROUND The impact of adjuvant radiation in patients with atypical meningioma remains poorly defined. We sought to determine the impact of adjuvant radiation therapy in this population. METHODS We identified 91 patients with World Health Organization grade II (atypical) meningioma managed at Dana-Farber/Brigham and Womens Cancer Center between 1997 and 2011. A propensity score model incorporating age at diagnosis, gender, Karnofsky performance status, tumor location, tumor size, reason for diagnosis, and era of treatment was constructed using logistic regression for the outcome of receipt versus nonreceipt of radiation therapy. Propensity scores were then used as continuous covariates in a Cox proportional hazards model to determine the adjusted impact of adjuvant radiation therapy on both local recurrence and the combined endpoint of use of salvage therapy and death due to progressive meningioma. RESULTS The median follow-up in patients without recurrent disease was 4.9 years. After adjustment for pertinent confounding variables, radiation therapy was associated with decreased local recurrence in those undergoing gross total resection (hazard ratio, 0.25; 95% CI, 0.07-0.96; P = .04). No differences in overall survival were seen in patients who did and did not receive radiation therapy. CONCLUSION Patients who have had a gross total resection of an atypical meningioma should be considered for adjuvant radiation therapy given the improvement in local control. Multicenter, prospective trials are required to definitively evaluate the potential impact of radiation therapy on survival in patients with atypical meningioma.


Glia | 2007

Altering DNA base excision repair: Use of nuclear and mitochondrial- targeted N-methylpurine DNA glycosylase to sensitize astroglia to chemotherapeutic agents

Jason F. Harrison; Mikael L. Rinne; Mark R. Kelley; Nadiya M. Druzhyna; Glenn L. Wilson; Susan P. LeDoux

Primary astrocyte cultures were used to investigate the modulation of DNA repair as a tool for sensitizing astrocytes to genotoxic agents. Base excision repair (BER) is the principal mechanism by which mammalian cells repair alkylation damage to DNA and involves the processing of relatively nontoxic DNA adducts through a series of cytotoxic intermediates during the course of restoring normal DNA integrity. An adenoviral expression system was employed to target high levels of the BER pathway initiator, N‐methylpurine glycosylase (MPG), to either the mitochondria or nucleus of primary astrocytes to test the hypothesis that an alteration in BER results in increased alkylation sensitivity. Increasing MPG activity significantly increased BER kinetics in both the mitochondria and nuclei. Although modulating MPG activity in mitochondria appeared to have little effect on alkylation sensitivity, increased nuclear MPG activity resulted in cell death in astrocyte cultures treated with methylnitrosourea (MNU). Caspase‐3 cleavage was not detected, thus indicating that these alkylation sensitive astrocytes do not undergo a typical programmed cell death in response to MNU. Astrocytes were found to express relatively high levels of antiapoptotic Bcl‐2 and Bcl‐XL and very low levels of proapoptotic Bad and Bid suggesting that the mitochondrial pathway of apoptosis may be blocked making astrocytes less vulnerable to proapoptotic stimuli compared with other cell types. Consequently, this unique characteristic of astrocytes may be responsible, in part, for resistance of astrocytomas to chemotherapeutic agents.


Neuro-oncology | 2015

Clinical implementation of integrated whole-genome copy number and mutation profiling for glioblastoma

Shakti Ramkissoon; Wenya Linda Bi; Steven E. Schumacher; Lori A. Ramkissoon; Sam Haidar; David Knoff; Adrian Dubuc; Loreal Brown; Margot Burns; Jane Cryan; Malak Abedalthagafi; Yun Jee Kang; Nikolaus Schultz; David A. Reardon; Eudocia Q. Lee; Mikael L. Rinne; Andrew D. Norden; Lakshmi Nayak; Sandra Ruland; Lisa Doherty; Debra C. LaFrankie; M.C. Horvath; Ayal A. Aizer; Andrea L. Russo; Nils D. Arvold; Elizabeth B. Claus; Ossama Al-Mefty; Mark D. Johnson; Alexandra J. Golby; Ian F. Dunn

BACKGROUND Multidimensional genotyping of formalin-fixed paraffin-embedded (FFPE) samples has the potential to improve diagnostics and clinical trials for brain tumors, but prospective use in the clinical setting is not yet routine. We report our experience with implementing a multiplexed copy number and mutation-testing program in a diagnostic laboratory certified by the Clinical Laboratory Improvement Amendments. METHODS We collected and analyzed clinical testing results from whole-genome array comparative genomic hybridization (OncoCopy) of 420 brain tumors, including 148 glioblastomas. Mass spectrometry-based mutation genotyping (OncoMap, 471 mutations) was performed on 86 glioblastomas. RESULTS OncoCopy was successful in 99% of samples for which sufficient DNA was obtained (n = 415). All clinically relevant loci for glioblastomas were detected, including amplifications (EGFR, PDGFRA, MET) and deletions (EGFRvIII, PTEN, 1p/19q). Glioblastoma patients ≤40 years old had distinct profiles compared with patients >40 years. OncoMap testing reliably identified mutations in IDH1, TP53, and PTEN. Seventy-seven glioblastoma patients enrolled on trials, of whom 51% participated in targeted therapeutic trials where multiplex data informed eligibility or outcomes. Data integration identified patients with complete tumor suppressor inactivation, albeit rarely (5% of patients) due to lack of whole-gene coverage in OncoMap. CONCLUSIONS Combined use of multiplexed copy number and mutation detection from FFPE samples in the clinical setting can efficiently replace singleton tests for clinical diagnosis and prognosis in most settings. Our results support incorporation of these assays into clinical trials as integral biomarkers and their potential to impact interpretation of results. Limited tumor suppressor variant capture by targeted genotyping highlights the need for whole-gene sequencing in glioblastoma.


British Journal of Haematology | 2016

Central nervous system involvement by Waldenström macroglobulinaemia (Bing-Neel syndrome): a multi-institutional retrospective study.

Jorge J. Castillo; Shirley D'Sa; Michael P. Lunn; Monique C. Minnema; Alessandra Tedeschi; Frederick Lansigan; M. Lia Palomba; Marzia Varettoni; Ramón García-Sanz; Lakshmi Nayak; Eudocia Q. Lee; Mikael L. Rinne; Andrew D. Norden; Irene M. Ghobrial; Steven P. Treon

Bing‐Neel syndrome (BNS) is a rare complication seen in patients with Waldenström macroglobulinaemia (WM), in which lymphoplasmacytic lymphoma cells colonize the central nervous system. In this retrospective multi‐centre study, we present the clinicopathological features, imaging findings, therapy, response and outcomes of 34 patients with BNS. The median time from WM diagnosis to BNS diagnosis was 3 years, 15% of patients were diagnosed with BNS at the time of WM diagnosis, and 22% of patients developed BNS when responding to active treatment for WM. Patients with BNS presented with variable clinical features including limb motor deficits, change in mental status and cranial nerve palsies. The diagnosis was made using a combination of cerebrospinal fluid cytology, flow cytometry and detection of the MYD88 L265 mutation, and magnetic resonance imaging. The estimated 3‐year overall survival rate was 59%. Of the survivors, 40% have evidence of pathological and/or radiological persistence of disease. Age older than 65 years, platelet count lower than 100 × 109/l, and treatment for WM prior to BNS diagnosis were associated with worse outcome. Exposure to rituximab for treatment of BNS was associated with a better outcome. Multi‐institutional collaboration is warranted to improve treatment and outcomes in patients with BNS.


JAMA Oncology | 2015

Rapid Intraoperative Molecular Characterization of Glioma

Ganesh M. Shankar; Joshua M. Francis; Mikael L. Rinne; Shakti Ramkissoon; Franklin W. Huang; Andrew S. Venteicher; Elliot H. Akama-Garren; Yun Jee Kang; Nina Lelic; James C. Kim; Loreal Brown; Sarah K. Charbonneau; Alexandra J. Golby; Chandra Sekhar Pedamallu; Mai P. Hoang; Ryan J. Sullivan; Andrew D. Cherniack; Levi A. Garraway; Anat Stemmer-Rachamimov; David A. Reardon; Patrick Y. Wen; Priscilla K. Brastianos; William T. Curry; Fred G. Barker; William C. Hahn; Brian V. Nahed; Keith L. Ligon; David N. Louis; Daniel P. Cahill; Matthew Meyerson

IMPORTANCE Conclusive intraoperative pathologic confirmation of diffuse infiltrative glioma guides the decision to pursue definitive neurosurgical resection. Establishing the intraoperative diagnosis by histologic analysis can be difficult in low-cellularity infiltrative gliomas. Therefore, we developed a rapid and sensitive genotyping assay to detect somatic single-nucleotide variants in the telomerase reverse transcriptase (TERT) promoter and isocitrate dehydrogenase 1 (IDH1). OBSERVATIONS This assay was applied to tissue samples from 190 patients with diffuse gliomas, including archived fixed and frozen specimens and tissue obtained intraoperatively. Results demonstrated 96% sensitivity (95% CI, 90%-99%) and 100% specificity (95% CI, 95%-100%) for World Health Organization grades II and III gliomas. In a series of live cases, glioma-defining mutations could be identified within 60 minutes, which could facilitate the diagnosis in an intraoperative timeframe. CONCLUSIONS AND RELEVANCE The genotyping method described herein can establish the diagnosis of low-cellularity tumors like glioma and could be adapted to the point-of-care diagnosis of other lesions that are similarly defined by highly recurrent somatic mutations.


Expert Opinion on Emerging Drugs | 2013

Update on bevacizumab and other angiogenesis inhibitors for brain cancer

Mikael L. Rinne; Eudocia Q. Lee; Lakshmi Nayak; Andrew D. Norden; Rameen Beroukhim; Patrick Y. Wen; David A. Reardon

Introduction: Primary and metastatic brain tumors remain a major challenge. The most common primary adult malignant brain tumor, glioblastoma (GBM), confers a dismal prognosis as does the development of CNS metastases for most systemic malignancies. Anti-angiogenic therapy has been a major clinical research focus in neuro-oncology over the past 5 years. Areas covered: Culmination of this work includes US FDA accelerated approval of bevacizumab for recurrent GBM and the completion of two placebo-controlled Phase III studies of bevacizumab for newly diagnosed GBM. A multitude of anti-angiogenics are in evaluation for neuro-oncology patients but none has thus far surpassed the therapeutic benefit of bevacizumab. Expert opinion: These agents demonstrate adequate safety and the majority of GBM patients derive benefit. Furthermore, their anti-permeability effect can substantially decrease tumor-associated edema leading to stable or improved neurologic function and quality of life. In particular, anti-angiogenics significantly prolong progression-free survival – a noteworthy achievement in the context of infiltrative and destructive brain tumors like GBM; however, in a manner analogous to other cancers, their impact on overall survival for GBM patients is modest at best. Despite substantial clinical research efforts, many fundamental questions regarding anti-angiogenic agents in brain tumor patients remain unanswered.

Collaboration


Dive into the Mikael L. Rinne's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian M. Alexander

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge