Network


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

Hotspot


Dive into the research topics where Chandralekha Halder is active.

Publication


Featured researches published by Chandralekha Halder.


The New England Journal of Medicine | 2015

Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors

Jeanette E. Eckel-Passow; Daniel H. Lachance; Annette M. Molinaro; Kyle M. Walsh; Paul A. Decker; Hugues Sicotte; Melike Pekmezci; Terri Rice; Matt L. Kosel; Ivan Smirnov; Gobinda Sarkar; Alissa Caron; Thomas M. Kollmeyer; Corinne Praska; Anisha R. Chada; Chandralekha Halder; Helen M. Hansen; Lucie McCoy; Paige M. Bracci; Roxanne Marshall; Shichun Zheng; Gerald F. Reis; Alexander R. Pico; Brian Patrick O’Neill; Jan C. Buckner; Caterina Giannini; Jason T. Huse; Arie Perry; Tarik Tihan; Mitchell S. Berger

BACKGROUND The prediction of clinical behavior, response to therapy, and outcome of infiltrative glioma is challenging. On the basis of previous studies of tumor biology, we defined five glioma molecular groups with the use of three alterations: mutations in the TERT promoter, mutations in IDH, and codeletion of chromosome arms 1p and 19q (1p/19q codeletion). We tested the hypothesis that within groups based on these features, tumors would have similar clinical variables, acquired somatic alterations, and germline variants. METHODS We scored tumors as negative or positive for each of these markers in 1087 gliomas and compared acquired alterations and patient characteristics among the five primary molecular groups. Using 11,590 controls, we assessed associations between these groups and known glioma germline variants. RESULTS Among 615 grade II or III gliomas, 29% had all three alterations (i.e., were triple-positive), 5% had TERT and IDH mutations, 45% had only IDH mutations, 7% were triple-negative, and 10% had only TERT mutations; 5% had other combinations. Among 472 grade IV gliomas, less than 1% were triple-positive, 2% had TERT and IDH mutations, 7% had only IDH mutations, 17% were triple-negative, and 74% had only TERT mutations. The mean age at diagnosis was lowest (37 years) among patients who had gliomas with only IDH mutations and was highest (59 years) among patients who had gliomas with only TERT mutations. The molecular groups were independently associated with overall survival among patients with grade II or III gliomas but not among patients with grade IV gliomas. The molecular groups were associated with specific germline variants. CONCLUSIONS Gliomas were classified into five principal groups on the basis of three tumor markers. The groups had different ages at onset, overall survival, and associations with germline variants, which implies that they are characterized by distinct mechanisms of pathogenesis. (Funded by the National Institutes of Health and others.).


Nature Genetics | 2009

Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility.

Margaret Wrensch; Robert B. Jenkins; Jeffrey S. Chang; Ru Fang Yeh; Yuanyuan Xiao; Paul A. Decker; Karla V. Ballman; Mitchel S. Berger; Jan C. Buckner; Susan M. Chang; Caterina Giannini; Chandralekha Halder; Thomas M. Kollmeyer; Matthew L. Kosel; Daniel H. Lachance; Lucie McCoy; Brian Patrick O'Neill; Joe Patoka; Alexander R. Pico; Michael D. Prados; Charles P. Quesenberry; Terri Rice; Amanda L. Rynearson; Ivan Smirnov; Tarik Tihan; Joseph L. Wiemels; Ping Yang; John K. Wiencke

The causes of glioblastoma and other gliomas remain obscure. To discover new candidate genes influencing glioma susceptibility, we conducted a principal component–adjusted genome-wide association study (GWAS) of 275,895 autosomal variants among 692 adult high-grade glioma cases (622 from the San Francisco Adult Glioma Study (AGS) and 70 from the Cancer Genome Atlas (TCGA)) and 3,992 controls (602 from AGS and 3,390 from Illumina iControlDB (iControls)). For replication, we analyzed the 13 SNPs with P < 10−6 using independent data from 176 high-grade glioma cases and 174 controls from the Mayo Clinic. On 9p21, rs1412829 near CDKN2B had discovery P = 3.4 × 10−8, replication P = 0.0038 and combined P = 1.85 × 10−10. On 20q13.3, rs6010620 intronic to RTEL1 had discovery P = 1.5 × 10−7, replication P = 0.00035 and combined P = 3.40 × 10−9. For both SNPs, the direction of association was the same in discovery and replication phases.


Nature Genetics | 2012

A low-frequency variant at 8q24.21 is strongly associated with risk of oligodendroglial tumors and astrocytomas with IDH1 or IDH2 mutation

Robert B. Jenkins; Yuanyuan Xiao; Hugues Sicotte; Paul A. Decker; Thomas M. Kollmeyer; Helen M. Hansen; Matthew L. Kosel; Shichun Zheng; Kyle M. Walsh; Terri Rice; Paige M. Bracci; Lucie McCoy; Ivan Smirnov; Joseph S. Patoka; George Hsuang; Joseph L. Wiemels; Tarik Tihan; Alexander R. Pico; Michael D. Prados; Susan M. Chang; Mitchel S. Berger; Alissa Caron; Stephanie R. Fink; Chandralekha Halder; Amanda L. Rynearson; Brooke L. Fridley; Jan C. Buckner; Brian Patrick O'Neill; Caterina Giannini; Daniel H. Lachance

Variants at 8q24.21 have been shown to be associated with glioma development. By means of tag SNP genotyping and imputation, pooled next-generation sequencing using long-range PCR and subsequent validation SNP genotyping, we identified seven low-frequency SNPs at 8q24.21 that were strongly associated with glioma risk (P = 1 × 10−25 to 1 × 10−14). The most strongly associated SNP, rs55705857, remained highly significant after individual adjustment for the other top six SNPs and two previously published SNPs. After stratifying by histological and tumor genetic subtype, the most significant associations of rs55705857 were with oligodendroglial tumors and gliomas with mutant IDH1 or IDH2 (odds ratio (OR) = 5.1, P = 1.1 × 10−31 and OR = 4.8, P = 6.6 × 10−22, respectively). Strong associations were observed for astrocytomas with mutated IDH1 or IDH2 (grades 2–4) (OR = 5.16–6.66, P = 4.7 × 10−12 to 2.2 × 10−8) but not for astrocytomas with wild-type IDH1 and IDH2 (smallest P = 0.26). The conserved sequence block that includes rs55705857 is consistently modeled as a microRNA.


Cancer Genetics and Cytogenetics | 2011

Distinct germ line polymorphisms underlie glioma morphologic heterogeneity

Robert B. Jenkins; Margaret Wrensch; Derek R. Johnson; Brooke L. Fridley; Paul A. Decker; Yuanyuan Xiao; Thomas M. Kollmeyer; Amanda L. Rynearson; Stephanie R. Fink; Terri Rice; Lucie McCoy; Chandralekha Halder; Matthew L. Kosel; Caterina Giannini; Tarik Tihan; Brian Patrick O’Neill; Daniel H. Lachance; Ping Yang; Joseph L. Wiemels; John K. Wiencke

Two recent genome-wide association studies reported that single nucleotide polymorphisms (SNPs) in (or near) TERT (5p15), CCDC26 (8q24), CDKN2A/B (9p21), PHLDB1 (11q23), and RTEL1 (20q13) are associated with infiltrating glioma. From these reports, it was not clear whether the single nucleotide polymorphism associations predispose to glioma in general or whether they are specific to certain glioma grades or morphologic subtypes. To identify hypothesized associations between susceptibility loci and tumor subtype, we genotyped two case-control groups composed of the spectrum of infiltrating glioma subtypes and stratified the analyses by type. We report that specific germ line polymorphisms are associated with different glioma subtypes. CCDC26 (8q24) region polymorphisms are strongly associated with oligodendroglial tumor risk (rs4295627, odds ratio [OR] = 2.05, P = 8.3 × 10(-11)) but not glioblastoma risk. The opposite is true of RTEL (20q13) region polymorphisms, which are significantly associated with glioblastoma (rs2297440, OR = 0.56, P = 4.6 × 10(-10)) but not oligodendroglial tumor. The SNPs in or near CCDC26 (8q24) are associated with oligodendroglial tumors regardless of combined 1p and 19q deletion status; however, the association is greatest for those with combined deletion (rs4295627, OR = 2.77, P = 2.6 × 10(-9)). These observations generate hypotheses concerning the possible mechanisms by which specific SNPs (or alterations in linkage disequilibrium with such SNPs) are associated with glioma development.


Cancer Research | 2010

Sleeping Beauty–Mediated Somatic Mutagenesis Implicates CSF1 in the Formation of High-Grade Astrocytomas

Aaron M. Bender; Lara S. Collier; Fausto J. Rodriguez; Christina Tieu; Jon D. Larson; Chandralekha Halder; Eric Mahlum; Thomas M. Kollmeyer; Keiko Akagi; Gobinda Sarkar; David A. Largaespada; Robert B. Jenkins

The Sleeping Beauty (SB) transposon system has been used as an insertional mutagenesis tool to identify novel cancer genes. To identify glioma-associated genes, we evaluated tumor formation in the brain tissue from 117 transgenic mice that had undergone constitutive SB-mediated transposition. Upon analysis, 21 samples (18%) contained neoplastic tissue with features of high-grade astrocytomas. These tumors expressed glial markers and were histologically similar to human glioma. Genomic DNA from SB-induced astrocytoma tissue was extracted and transposon insertion sites were identified. Insertions in the growth factor gene Csf1 were found in 13 of the 21 tumors (62%), clustered in introns 5 and 8. Using reverse transcription-PCR, we documented increased Csf1 RNAs in tumor versus adjacent normal tissue, with the identification of transposon-terminated Csf1 mRNAs in astrocytomas with SB insertions in intron 8. Analysis of human glioblastomas revealed increased levels of Csf1 RNA and protein. Together, these results indicate that SB-insertional mutagenesis can identify high-grade astrocytoma-associated genes and they imply an important role for CSF1 in the development of these tumors.


Clinical Cancer Research | 2006

High WT1 expression is associated with very poor survival of patients with osteogenic sarcoma metastasis.

Alok Srivastava; Bruno Fuchs; Kunbo Zhang; Ming Ruan; Chandralekha Halder; Eric Mahlum; Kristin Weber; Mark E. Bolander; Gobinda Sarkar

Purpose: Although metastasis is the primary determinant of poor survival of patients with osteogenic sarcoma, some patients live much longer than others, indicating metastatic heterogeneity underlying survival outcome. The purpose of the investigation was to identify genes underlying survival outcome of patients with osteogenic sarcoma metastasis. Experimental Design: We have used microarray to first compare mRNA expression between normal bone and osteogenic sarcoma specimens, identified genes overexpressed in osteogenic sarcoma, and compared expression of the selected gene between a poorly metastatic (SAOS) and two highly metastatic cell lines (LM8 and 143B). Finally, expression of the selected gene was assessed by immunostaining of osteogenic sarcoma samples with known survival outcome. Results: Microarray analysis revealed 5.3-fold more expression of WT1 mRNA in osteogenic sarcoma compared with normal bone and >2-fold overexpression in 143B and LM8 cells compared with SAOS. Furthermore, WT1 mRNA was absent in normal bone (10 of 10) by reverse transcription-PCR but present in osteogenic sarcoma–derived cell lines (5 of 8). One hundred percent (42 of 42) of low-grade osteogenic sarcoma specimens expressed no WT1 as determined by immunostaining; however, 24% (12 of 49) of the high-grade specimens showed intense staining. Mean survival of patients with high-grade metastatic osteogenic sarcoma but low WT1 staining (27 of 37) was 96.5 ± 129.3 months, whereas mean survival of patients with high-grade metastatic osteogenic sarcoma having intense staining (10 of 37) was 18.3 ± 12.3 months (P > 0.0143). All splice variants of WT1 mRNA, including a hitherto unknown variant (lacking exons 4 and 5), were found to be expressed in osteogenic sarcoma. Conclusion: WT1 seems to be associated with very poor survival of patients with osteogenic sarcoma metastasis.


Gene | 2007

High expression of tumor endothelial marker 7 is associated with metastasis and poor survival of patients with osteogenic sarcoma

Bruno Fuchs; Eric Mahlum; Chandralekha Halder; Avudaippan Maran; Michael J. Yaszemski; Beata Bode; Mark E. Bolander; Gobinda Sarkar


Analytical Biochemistry | 2007

Engineering a noncarrier to a highly efficient carrier peptide for noncovalently delivering biologically active proteins into human cells.

Eric Mahlum; Deendayal Mandal; Chandralekha Halder; Avudaiappan Maran; Michael J. Yaszemski; Robert B. Jenkins; Mark E. Bolander; Gobinda Sarkar


Anticancer Research | 2009

Preferential Expression of the Secreted and Membrane Forms of Tumor Endothelial Marker 7 Transcripts in Osteosarcoma

Chandralekha Halder; Christian Ossendorf; Avudaiappan Maran; Michael J. Yaszemski; Mark E. Bolander; Bruno Fuchs; Gobinda Sarkar


Neurosurgery | 2017

156 Small Terminal Deletions/Duplications and Alternative Lengthening of Telomeres are Co-Occur in IDH Mutation Only Gliomas

Desmond A. Brown; Seiji Yamada; Thomas M. Kollmeyer; Paul A. Decker; Matthew L. Kosel; Gobinda Sarkar; Alissa Caron; Chandralekha Halder; Vanessa Y Ruiz; Benjamin R. Kipp; Jesse S. Voss; Caterina Giannini; Robert B. Jenkins

Collaboration


Dive into the Chandralekha Halder'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

Terri Rice

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge