Network


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

Hotspot


Dive into the research topics where Mari Mino-Kenudson is active.

Publication


Featured researches published by Mari Mino-Kenudson.


The New England Journal of Medicine | 2010

Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer.

Eunice L. Kwak; Yung-Jue Bang; D. Ross Camidge; Alice T. Shaw; Benjamin Solomon; Robert G. Maki; Sai-Hong Ignatius Ou; Bruce J. Dezube; Pasi A. Jänne; Daniel B. Costa; Marileila Varella-Garcia; Woo-Ho Kim; Thomas J. Lynch; Panos Fidias; Hannah Stubbs; Jeffrey A. Engelman; Lecia V. Sequist; Weiwei Tan; Leena Gandhi; Mari Mino-Kenudson; Greg C. Wei; S. Martin Shreeve; Mark J. Ratain; Jeffrey Settleman; James G. Christensen; Daniel A. Haber; Keith D. Wilner; Ravi Salgia; Geoffrey I. Shapiro; Jeffrey W. Clark

BACKGROUND Oncogenic fusion genes consisting of EML4 and anaplastic lymphoma kinase (ALK) are present in a subgroup of non-small-cell lung cancers, representing 2 to 7% of such tumors. We explored the therapeutic efficacy of inhibiting ALK in such tumors in an early-phase clinical trial of crizotinib (PF-02341066), an orally available small-molecule inhibitor of the ALK tyrosine kinase. METHODS After screening tumor samples from approximately 1500 patients with non-small-cell lung cancer for the presence of ALK rearrangements, we identified 82 patients with advanced ALK-positive disease who were eligible for the clinical trial. Most of the patients had received previous treatment. These patients were enrolled in an expanded cohort study instituted after phase 1 dose escalation had established a recommended crizotinib dose of 250 mg twice daily in 28-day cycles. Patients were assessed for adverse events and response to therapy. RESULTS Patients with ALK rearrangements tended to be younger than those without the rearrangements, and most of the patients had little or no exposure to tobacco and had adenocarcinomas. At a mean treatment duration of 6.4 months, the overall response rate was 57% (47 of 82 patients, with 46 confirmed partial responses and 1 confirmed complete response); 27 patients (33%) had stable disease. A total of 63 of 82 patients (77%) were continuing to receive crizotinib at the time of data cutoff, and the estimated probability of 6-month progression-free survival was 72%, with no median for the study reached. The drug resulted in grade 1 or 2 (mild) gastrointestinal side effects. CONCLUSIONS The inhibition of ALK in lung tumors with the ALK rearrangement resulted in tumor shrinkage or stable disease in most patients. (Funded by Pfizer and others; ClinicalTrials.gov number, NCT00585195.).


Science Translational Medicine | 2011

Genotypic and Histological Evolution of Lung Cancers Acquiring Resistance to EGFR Inhibitors

Lecia V. Sequist; Belinda A. Waltman; Dora Dias-Santagata; Subba R. Digumarthy; Alexa B. Turke; P. Fidias; Kristin Bergethon; Alice T. Shaw; Scott N. Gettinger; Arjola K. Cosper; Sara Akhavanfard; Rebecca S. Heist; Jennifer S. Temel; James G. Christensen; John Wain; Thomas J. Lynch; Kathy Vernovsky; Eugene J. Mark; Anthony John Iafrate; Mari Mino-Kenudson; J. A. Engelman

Lung cancers undergo dynamic genetic and histological changes upon developing resistance to EGFR inhibitors. The Shifting Sands of Lung Cancer Lung cancer is the leading cause of death globally and has proven very difficult to treat. The development almost a decade ago of tyrosine kinase inhibitors that specifically block the epidermal growth factor receptor (EGFR), which is switched on in many lung cancers, provided hope that targeted therapies would finally combat this deadly disease. However, only a certain subpopulation of lung cancer patients carrying specific activating mutations in EGFR responded clinically to EGFR inhibitors, and even among these patients, resistance to the inhibitor emerged within 12 months. To better understand how lung cancers develop drug resistance, Sequist and colleagues undertook a comprehensive genetic and histological analysis of 37 patients with non–small cell lung cancer (NSCLC), and they made some surprising discoveries. In an effort to understand the exact mechanism underscoring the acquisition of drug resistance in NSCLC patients treated with EGFR inhibitors, the investigators analyzed tumor biopsies from patients at the time they acquired resistance. All of the lung cancer patients retained their original activating EGFR mutations, but some patients had acquired another mutation in EGFR (T790M), which interferes with binding of the drug to the receptor, rendering the tumors resistant. Meanwhile, another group of patients became resistant because they developed amplification of a gene encoding the MET tyrosine kinase receptor, which, like EGFR, drives cell growth. Yet other patients acquired drug resistance mechanisms that had not been reported before including amplification of the EGFR gene itself and mutations in the PIK3CA gene (which encodes a subunit of the signaling molecule phosphatidylinositol 3-kinase). In addition, the authors observed that a few lung cancers transitioned from an epithelial cell morphology to a mesenchymal cell–like appearance, which is associated with a more aggressive type of tumor. In five patients, the authors discovered another type of transition that was even more surprising: the conversion of NSCLCs into small cell lung cancers (SCLCs), which are easier to treat. Indeed, these five patients responded well to the typical chemotherapy regimen used to treat SCLCs. To study the evolution of lung tumors in patients over the course of their disease, the investigators took serial biopsies from three lung cancer patients over 2 years. They found that when the patients acquired drug resistance and were then taken off the EGFR inhibitor, they lost the resistance mutations and their tumors once again became sensitive to treatment by either the same or a different EGFR inhibitor. The detailed genetic and histological analysis by Sequist and colleagues provides new insights into the shifting sands of drug resistance evolution in lung cancers and suggests that serial biopsies may be essential in the quest to reverse or even prevent the development of drug resistance. Lung cancers harboring mutations in the epidermal growth factor receptor (EGFR) respond to EGFR tyrosine kinase inhibitors, but drug resistance invariably emerges. To elucidate mechanisms of acquired drug resistance, we performed systematic genetic and histological analyses of tumor biopsies from 37 patients with drug-resistant non–small cell lung cancers (NSCLCs) carrying EGFR mutations. All drug-resistant tumors retained their original activating EGFR mutations, and some acquired known mechanisms of resistance including the EGFR T790M mutation or MET gene amplification. Some resistant cancers showed unexpected genetic changes including EGFR amplification and mutations in the PIK3CA gene, whereas others underwent a pronounced epithelial-to-mesenchymal transition. Surprisingly, five resistant tumors (14%) transformed from NSCLC into small cell lung cancer (SCLC) and were sensitive to standard SCLC treatments. In three patients, serial biopsies revealed that genetic mechanisms of resistance were lost in the absence of the continued selective pressure of EGFR inhibitor treatment, and such cancers were sensitive to a second round of treatment with EGFR inhibitors. Collectively, these results deepen our understanding of resistance to EGFR inhibitors and underscore the importance of repeatedly assessing cancers throughout the course of the disease.


Journal of Clinical Oncology | 2009

Clinical Features and Outcome of Patients With Non–Small-Cell Lung Cancer Who Harbor EML4-ALK

Alice T. Shaw; Beow Y. Yeap; Mari Mino-Kenudson; Subba R. Digumarthy; Daniel B. Costa; Rebecca S. Heist; Benjamin Solomon; Hannah Stubbs; Sonal Admane; Ultan McDermott; Jeffrey Settleman; Susumu Kobayashi; Eugene J. Mark; Scott J. Rodig; Lucian R. Chirieac; Eunice L. Kwak; Thomas J. Lynch; A. John Iafrate

PURPOSE The EML4-ALK fusion oncogene represents a novel molecular target in a small subset of non-small-cell lung cancers (NSCLC). To aid in identification and treatment of these patients, we examined the clinical characteristics and treatment outcomes of patients who had NSCLC with and without EML4-ALK. PATIENTS AND METHODS Patients with NSCLC were selected for genetic screening on the basis of two or more of the following characteristics: female sex, Asian ethnicity, never/light smoking history, and adenocarcinoma histology. EML4-ALK was identified by using fluorescent in situ hybridization for ALK rearrangements and was confirmed by immunohistochemistry for ALK expression. EGFR and KRAS mutations were determined by DNA sequencing. RESULTS Of 141 tumors screened, 19 (13%) were EML4-ALK mutant, 31 (22%) were EGFR mutant, and 91 (65%) were wild type (WT/WT) for both ALK and EGFR. Compared with the EGFR mutant and WT/WT cohorts, patients with EML4-ALK mutant tumors were significantly younger (P < .001 and P = .005) and were more likely to be men (P = .036 and P = .039). Patients with EML4-ALK-positive tumors, like patients who harbored EGFR mutations, also were more likely to be never/light smokers compared with patients in the WT/WT cohort (P < .001). Eighteen of the 19 EML4-ALK tumors were adenocarcinomas, predominantly the signet ring cell subtype. Among patients with metastatic disease, EML4-ALK positivity was associated with resistance to EGFR tyrosine kinase inhibitors (TKIs). Patients in the EML4-ALK cohort and the WT/WT cohort showed similar response rates to platinum-based combination chemotherapy and no difference in overall survival. CONCLUSION EML4-ALK defines a molecular subset of NSCLC with distinct clinical characteristics. Patients who harbor this mutation do not benefit from EGFR TKIs and should be directed to trials of ALK-targeted agents.


Journal of Clinical Oncology | 2006

Chemotherapy Regimen Predicts Steatohepatitis and an Increase in 90-Day Mortality After Surgery for Hepatic Colorectal Metastases

Jean Nicolas Vauthey; Timothy M. Pawlik; Dario Ribero; Tsung Teh Wu; Daria Zorzi; Paulo M. Hoff; Henry Q. Xiong; Cathy Eng; Gregory Y. Lauwers; Mari Mino-Kenudson; Mauro Risio; Andrea Muratore; Lorenzo Capussotti; Steven A. Curley; Eddie K. Abdalla

PURPOSE Chemotherapy before resection of hepatic colorectal metastases (CRM) may cause hepatic injury and affect postoperative outcome. PATIENTS AND METHODS Four hundred six patients underwent hepatic resection of CRM between 1992 and 2005. Pathologic review of the nontumorous liver was performed using established criteria for steatosis, steatohepatitis, and sinusoidal injury. The effect of chemotherapy and liver injury on perioperative outcome was analyzed. RESULTS One hundred fifty-eight patients (38.9%) received no preoperative chemotherapy, whereas 248 patients (61.1%) did. The median duration of chemotherapy was 16 weeks (range, 2 to 70 weeks). Chemotherapy consisted of fluoropyrimidine-based regimens (fluorouracil [FU] alone, 15.5%; irinotecan plus FU, 23.1%; and oxaliplatin plus FU, 19.5%) and other therapy (3.0%). On pathologic analysis, 36 patients (8.9%) had steatosis, 34 (8.4%) had steatohepatitis, and 22 (5.4%) had sinusoidal dilation. Oxaliplatin was associated with sinusoidal dilation compared with no chemotherapy (18.9% v 1.9%, respectively; P < .001; odds ratio [OR] = 8.3; 95% CI, 2.9 to 23.6). In contrast, irinotecan was associated with steatohepatitis compared with no chemotherapy (20.2% v 4.4%, respectively; P < .001; OR = 5.4; 95% CI, 2.2 to 13.5). Patients with steatohepatitis had an increased 90-day mortality compared with patients who did not have steatohepatitis (14.7% v 1.6%, respectively; P = .001; OR = 10.5; 95% CI, 2.0 to 36.4). CONCLUSION Steatohepatitis is associated with an increased 90-day mortality after hepatic surgery. In patients with hepatic CRM, the chemotherapy regimen should be carefully considered because the risk of hepatotoxicity is significant.


Journal of Clinical Oncology | 2012

ROS1 Rearrangements Define a Unique Molecular Class of Lung Cancers

Kristin Bergethon; Alice T. Shaw; Sai-Hong Ignatius Ou; Ryohei Katayama; Christine M. Lovly; Nerina T. McDonald; Pierre P. Massion; Christina Siwak-Tapp; Adriana Gonzalez; Rong Fang; Eugene J. Mark; Julie M. Batten; Haiquan Chen; Keith D. Wilner; Eunice L. Kwak; Jeffrey W. Clark; David P. Carbone; Hongbin Ji; Jeffrey A. Engelman; Mari Mino-Kenudson; William Pao; A. John Iafrate

PURPOSE Chromosomal rearrangements involving the ROS1 receptor tyrosine kinase gene have recently been described in a subset of non-small-cell lung cancers (NSCLCs). Because little is known about these tumors, we examined the clinical characteristics and treatment outcomes of patients with NSCLC with ROS1 rearrangement. PATIENTS AND METHODS Using a ROS1 fluorescent in situ hybridization (FISH) assay, we screened 1,073 patients with NSCLC and correlated ROS1 rearrangement status with clinical characteristics, overall survival, and when available, ALK rearrangement status. In vitro studies assessed the responsiveness of cells with ROS1 rearrangement to the tyrosine kinase inhibitor crizotinib. The clinical response of one patient with ROS1-rearranged NSCLC to crizotinib was investigated as part of an expanded phase I cohort. RESULTS Of 1,073 tumors screened, 18 (1.7%) were ROS1 rearranged by FISH, and 31 (2.9%) were ALK rearranged. Compared with the ROS1-negative group, patients with ROS1 rearrangements were significantly younger and more likely to be never-smokers (each P < .001). All of the ROS1-positive tumors were adenocarcinomas, with a tendency toward higher grade. ROS1-positive and -negative groups showed no difference in overall survival. The HCC78 ROS1-rearranged NSCLC cell line and 293 cells transfected with CD74-ROS1 showed evidence of sensitivity to crizotinib. The patient treated with crizotinib showed tumor shrinkage, with a near complete response. CONCLUSION ROS1 rearrangement defines a molecular subset of NSCLC with distinct clinical characteristics that are similar to those observed in patients with ALK-rearranged NSCLC. Crizotinib shows in vitro activity and early evidence of clinical activity in ROS1-rearranged NSCLC.


Pancreas | 2011

International Consensus Diagnostic Criteria for Autoimmune Pancreatitis: Guidelines of the International Association of Pancreatology

Tooru Shimosegawa; Suresh T. Chari; Luca Frulloni; Terumi Kamisawa; Shigeyuki Kawa; Mari Mino-Kenudson; Myung-Hwan Kim; Günter Klöppel; Markus M. Lerch; Matthias Löhr; Kenji Notohara; Kazuichi Okazaki; Alexander Schneider; Lizhi Zhang

Objectives: To achieve the goal of developing international consensus diagnostic criteria (ICDC) for autoimmune pancreatitis (AIP). Methods: An international panel of experts met during the 14th Congress of the International Association of Pancreatology held in Fukuoka, Japan, from July 11 through 13, 2010. The proposed criteria represent a consensus opinion of the working group. Results: Autoimmune pancreatitis was classified into types 1 and 2. The ICDC used 5 cardinal features of AIP, namely, imaging of pancreatic parenchyma and duct, serology, other organ involvement, pancreatic histology, and an optional criterion of response to steroid therapy. Each feature was categorized as level 1 and 2 findings depending on the diagnostic reliability. The diagnosis of type 1 and type 2 AIP can be definitive or probable, and in some cases, the distinction between the subtypes may not be possible (AIP-not otherwise specified). Conclusions: The ICDC for AIP were developed based on the agreement of an international panel of experts in the hope that they will promote worldwide recognition of AIP. The categorization of AIP into types 1 and 2 should be helpful for further clarification of the clinical features, pathogenesis, and natural history of these diseases.


Nature | 2011

Functional genomics reveal that the serine synthesis pathway is essential in breast cancer

Richard Possemato; Kevin Marks; Yoav D. Shaul; Michael E. Pacold; Dohoon Kim; Kivanc Birsoy; Shalini Sethumadhavan; Hin-Koon Woo; Hyun Gyung Jang; Abhishek K. Jha; Walter W. Chen; Francesca G. Barrett; Nicolas Stransky; Zhi-Yang Tsun; Glenn S. Cowley; Jordi Barretina; Nada Y. Kalaany; Peggy P. Hsu; Kathleen Ottina; Albert M. Chan; Bingbing Yuan; Levi A. Garraway; David E. Root; Mari Mino-Kenudson; Elena F. Brachtel; Edward M. Driggers; David M. Sabatini

Cancer cells adapt their metabolic processes to drive macromolecular biosynthesis for rapid cell growth and proliferation (1,2). RNAi-based loss of function screening has proven powerful for the identification of novel and interesting cancer targets, and recent studies have used this technology in vivo to identify novel tumor suppressor genes (3). Here, we developed a method for identifying novel cancer targets via negative selection RNAi screening in solid tumours. Using this method, we screened a set of metabolic genes associated with aggressive breast cancer and stemness to identify those required for in vivo tumourigenesis. Among the genes identified, phosphoglycerate dehydrogenase (PHGDH) is in a genomic region of recurrent copy number gain in breast cancer and PHGDH protein levels are elevated in 70% of ER-negative breast cancers. PHGDH catalyzes the first step in the serine biosynthesis pathway, and breast cancer cells with high PHGDH expression have elevations in serine synthesis flux. Suppression of PHGDH in cell lines with elevated PHGDH expression, but not those without, causes a strong decrease in cell proliferation and a reduction in serine synthesis. We find that PHGDH suppression does not affect intracellular serine levels, but causes a drop in the levels of alpha-ketoglutarate, another output of the pathway and a TCA cycle intermediate. In cells with high PHGDH expression, the serine synthesis pathway contributes approximately 50% of the total anaplerotic flux of glutamine into the TCA cycle. These results reveal that certain breast cancers are dependent upon increased serine pathway flux caused by PHGDH over-expression and demonstrate the utility of in vivo negative selection RNAi screens for finding potential anticancer targets.


Clinical Cancer Research | 2009

Unique Clinicopathologic Features Characterize ALK-Rearranged Lung Adenocarcinoma in the Western Population

Scott J. Rodig; Mari Mino-Kenudson; Sanja Dacic; Beow Y. Yeap; Alice T. Shaw; Justine A. Barletta; Hannah Stubbs; Kenneth Law; Neal I. Lindeman; Eugene J. Mark; Pasi A. Jänne; Thomas R. Lynch; Bruce E. Johnson; Anthony John Iafrate; Lucian R. Chirieac

Purpose: The anaplastic large cell kinase gene (ALK) is rearranged in ∼5% of lung adenocarcinomas within the Asian population. We evaluated the incidence and the characteristics of ALK-rearranged lung adenocarcinomas within the western population and the optimal diagnostic modality to detect ALK rearrangements in routine clinical practice. Experimental Design: We tested 358 lung adenocarcinomas from three institutions for ALK rearrangements by fluorescent in situ hybridization (FISH) and immunohistochemistry with and without tyramide amplification. The clinicopathologic characteristics of tumors with and without ALK rearrangements were compared. Results: We identified 20 (5.6%) lung adenocarcinomas with ALK rearrangements within our cohort of western patients. ALK rearrangement was associated with younger age (P = 0.0002), never smoking (P < 0.0001), advanced clinical stage (P = 0.0001), and a solid histology with signet-ring cells (P < 0.0001). ALK rearrangement was identified by FISH in 95% of cases and immunohistochemistry with and without tyramide amplification in 80% and 40% of cases, respectively, but neither FISH nor immunohistochemistry alone detected all cases with ALK rearrangement on initial screening. None of the ALK-rearranged tumors harbored coexisting EGFR mutations. Conclusions: Lung adenocarcinomas with ALK rearrangements are uncommon in the western population and represent a distinct entity of carcinomas with unique characteristics. For suspected cases, dual diagnostic testing, with FISH and immunohistochemistry, should be considered to accurately identify lung adenocarcinomas with ALK rearrangement. (Clin Cancer Res 2009;15(16):5216–23)


The Journal of Molecular Diagnostics | 2005

Sensitive Sequencing Method for KRAS Mutation Detection by Pyrosequencing

Shuji Ogino; Takako Kawasaki; Mohan Brahmandam; Liying Yan; Mami Cantor; Chungdak Namgyal; Mari Mino-Kenudson; Gregory Y. Lauwers; Massimo Loda; Charles S. Fuchs

Both benign and malignant tumors represent heterogenous tissue containing tumor cells and non-neoplastic mesenchymal and inflammatory cells. To detect a minority of mutant KRAS alleles among abundant wild-type alleles, we developed a sensitive DNA sequencing assay using Pyrosequencing, ie, nucleotide extension sequencing with an allele quantification capability. We designed our Pyrosequencing assay for use with whole-genome-amplified DNA from paraffin-embedded tissue. Assessing various mixtures of DNA from mutant KRAS cell lines and DNA from a wild-type KRAS cell line, we found that mutation detection rates for Pyrosequencing were superior to dideoxy sequencing. In addition, Pyrosequencing proved superior to dideoxy sequencing in the detection of KRAS mutations from DNA mixtures of paraffin-embedded colon cancer and normal tissue as well as from paraffin-embedded pancreatic cancers. Quantification of mutant alleles by Pyrosequencing was precise and useful for assay validation, monitoring, and quality assurance. Our Pyrosequencing method is simple, robust, and sensitive, with a detection limit of approximately 5% mutant alleles. It is particularly useful for tumors containing abundant non-neoplastic cells. In addition, the applicability of this assay for DNA amplified by whole-genome amplification technique provides an expanded source of DNA for large-scale studies.


Cancer Discovery | 2012

EGFR-mediated re-activation of MAPK signaling contributes to insensitivity of BRAF mutant colorectal cancers to RAF inhibition with vemurafenib

Ryan B. Corcoran; Hiromichi Ebi; Alexa B. Turke; Erin M. Coffee; Michiya Nishino; Alexandria P. Cogdill; Ronald D. Brown; Patricia Della Pelle; Dora Dias-Santagata; Kenneth E. Hung; Keith T. Flaherty; Adriano Piris; Jennifer A. Wargo; Jeffrey Settleman; Mari Mino-Kenudson; Jeffrey A. Engelman

UNLABELLED BRAF mutations occur in 10-15% of colorectal cancers (CRCs) and confer adverse outcome. While RAF inhibitors such as vemurafenib (PLX4032) have proven effective in BRAF mutant melanoma, they are surprisingly ineffective in BRAF mutant CRCs, and the reason for this disparity remains unclear. Compared to BRAF mutant melanoma cells, BRAF mutant CRC cells were less sensitive to vemurafenib, and P-ERK suppression was not sustained in response to treatment. Although transient inhibition of phospho-ERK by vemurafenib was observed in CRC, rapid ERK re-activation occurred through EGFR-mediated activation of RAS and CRAF. BRAF mutant CRCs expressed higher levels of phospho-EGFR than BRAF mutant melanomas, suggesting that CRCs are specifically poised for EGFR-mediated resistance. Combined RAF and EGFR inhibition blocked reactivation of MAPK signaling in BRAF mutant CRC cells and markedly improved efficacy in vitro and in vivo. These findings support evaluation of combined RAF and EGFR inhibition in BRAF mutant CRC patients. SIGNIFICANCE BRAF valine 600 (V600) mutations occur in 10% to 15% of colorectal cancers, yet these tumors show a surprisingly low clinical response rate (~5%) to selective RAF inhibitors such as vemurafenib, which have produced dramatic response rates (60%–80%) in melanomas harboring the identical BRAF V600 mutation. We found that EGFR-mediated MAPK pathway reactivation leads to resistance to vemurafenib in BRAF-mutant colorectal cancers and that combined RAF and EGFR inhibition can lead to sustained MAPK pathway suppression and improved efficacy in vitro and in tumor xenografts.

Collaboration


Dive into the Mari Mino-Kenudson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gregory Y. Lauwers

University of South Florida

View shared research outputs
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
Researchain Logo
Decentralizing Knowledge