39 results on '"Balsara B"'
Search Results
2. LBA47 A phase II study evaluating tipifarnib in mHRAS, recurrent or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) (AIM-HN study)
- Author
-
Ho, A.L., Adkins, D.R., Hanna, G.J., Bruce, J., Ahn, M-J., Iglesias Docampo, L., Kang, H., Wong, D.J., Psyrri, A., Gillison, M., Braña, I., Liu, Y.C., Hsieh, C-Y., Hong, M.H., Zhang, Z., Balsara, B., Saunders, A., Gasco Hernandez, A., Dale, S., and Haddad, R.
- Published
- 2023
- Full Text
- View/download PDF
3. Detection of chromosomal aberrations by comparative genomic hybridization during transformation of human breast epithelial cells in vitro
- Author
-
Balogh, G., primary, Russo, I., additional, Balsara, B., additional, and Russo, J., additional
- Published
- 2006
- Full Text
- View/download PDF
4. Frequent activation of AKT in non-small cell lung carcinomas and preneoplastic bronchial lesions
- Author
-
Balsara, B. R., primary
- Published
- 2004
- Full Text
- View/download PDF
5. Neurofibromatosis 2 and malignant mesothelioma
- Author
-
Baser, M. E., primary, De Rienzo, A., additional, Altomare, D., additional, Balsara, B. R., additional, Hedrick, N. M., additional, Gutmann, D. H., additional, Pitts, L. H., additional, Jackler, R. K., additional, and Testa, J. R., additional
- Published
- 2002
- Full Text
- View/download PDF
6. Functional roles of chromosomes 11 and 17 in the transformation of human breast epithelial cells in vitro.
- Author
-
Yang, X, primary, Tahin, Q, additional, Hu, Y F, additional, Russo, I H, additional, Balsara, B R, additional, Mihaila, D, additional, Slater, C, additional, Barrett, J C, additional, and Russo, J, additional
- Published
- 1999
- Full Text
- View/download PDF
7. Characterization of MOAT-C and MOAT-D, New Members of the MRP/cMOAT Subfamily of Transporter Proteins
- Author
-
Belinsky, M. G., primary, Bain, L. J., additional, Balsara, B. B., additional, Testa, J. R., additional, and Kruh, G. D., additional
- Published
- 1998
- Full Text
- View/download PDF
8. Rhabdomyosarcoma
- Author
-
Gladstone, B., primary, Parikh, P.M., additional, Balsara, B., additional, Kadam, P.R., additional, Rao, S.R., additional, Nair, C.N., additional, Jambekar, N.A., additional, and Advani, S.H., additional
- Published
- 1993
- Full Text
- View/download PDF
9. T‐lymphoid blast crisis in chronic myeloid leukemia
- Author
-
Advani, S. H., primary, Malhotra, H., additional, Kadam, P. R., additional, Iyer, R. S., additional, Nanjangud, G., additional, Balsara, B., additional, Saikia, T., additional, Gopal, R., additional, and Nair, C. N., additional
- Published
- 1991
- Full Text
- View/download PDF
10. Advances in the Analysis of Chromosome Alterations in Human Lung Carcinomas
- Author
-
Testa, J. R., Liu, Z., Feder, M., Bell, D. W., Balsara, B., Cheng, Jin Quan, and Taguchi, T.
- Published
- 1997
- Full Text
- View/download PDF
11. Rhabdomyosarcoma: A cytogenetically interesting case report
- Author
-
Gladstone, B., Parikh, P.M., Balsara, B., Kadam, P.R., Rao, S.R., Nair, C.N., Jambekar, N.A., and Advani, S.H.
- Published
- 1993
- Full Text
- View/download PDF
12. Translocation t(1;21)(q21;p11) with Trisomies X and 1q in a Diffuse Large B-Cell Lymphoma
- Author
-
Gladstone, B. R., Kadam, P. R., Balsara, B., and Gopal, R.
- Published
- 1994
- Full Text
- View/download PDF
13. HRAS Mutations Define a Distinct Subgroup in Head and Neck Squamous Cell Carcinoma.
- Author
-
Coleman N, Marcelo KL, Hopkins JF, Khan NI, Du R, Hong L, Park E, Balsara B, Leoni M, Pickering C, Myers J, Heymach J, Albacker LA, Hong D, Gillison M, and Le X
- Subjects
- Aged, Humans, Male, Middle Aged, Mutation, Neoplasm Recurrence, Local, Proto-Oncogene Proteins p21(ras) genetics, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms genetics, Squamous Cell Carcinoma of Head and Neck drug therapy, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Purpose: In head and neck squamous cell carcinoma (HNSCC), HRAS mutation is a new actionable oncogene driver. We aimed to evaluate HRAS mutational variants, comutation profile, and survival outcomes of this molecularly defined population., Methods: We leveraged four deidentified patient data sets with HRAS -mutant HNSCC, MD Anderson Cancer Center, Kura Oncology, Inc trial, Foundation Medicine, and American Association for Cancer Research GENIE v.12. Patient demographic information and clinical courses were extracted, when available, in addition to HRAS mutation type and co-occurring mutations. Survival outcomes were analyzed (Kaplan-Meier method)., Results: Two hundred forty-nine patients with HRAS -mutant HNSCC were identified from the four data sets. Median age ranged from 55 to 65 years, with a higher frequency in male patients (64%); the majority of HRAS -mutant HNSCC occurred in human papillomavirus-negative HNSCC. HRAS mutation patterns were similar across data sets; G12S was the most common (29%). Treatment responses to tipifarnib were not codon-specific. Compared with wild-type, significantly co-occurring mutations with HRAS were Casp8 (Fisher's exact test, P < .00013), TERT ( P < .0085), and NOTCH1 ( P < .00013). Analysis of clinical courses from the MD Anderson Cancer Center and Kura Oncology, Inc data sets demonstrated poor clinical outcomes with a high rate of recurrence following primary definitive treatment (50%-67% relapse < 6 months) and short disease-free survival (4.0 months; 95% CI, 1.0 to 36.0) and overall survival (OS; 15.0 months; 95% CI, 6.0 to 52.0). Use of tipifarnib in this data set demonstrated improved OS (25.5 months; 95% CI, 18.0 to 48.0)., Conclusion: Oncogenic mutations in HRAS occur in 3%-4% of HNSCC, with G12S being the most frequent. Without targeted therapy, patients with HRAS -mutant HNSCC had poor clinic outcomes; observable trend toward improvement in OS has been noted in cohorts receiving treatments such as tipifarnib. The comutation pattern of HRAS -mutant in HNSCC is distinct, which may provide insight to future therapeutic combination strategies.
- Published
- 2023
- Full Text
- View/download PDF
14. Tipifarnib in Head and Neck Squamous Cell Carcinoma With HRAS Mutations.
- Author
-
Ho AL, Brana I, Haddad R, Bauman J, Bible K, Oosting S, Wong DJ, Ahn MJ, Boni V, Even C, Fayette J, Flor MJ, Harrington K, Kim SB, Licitra L, Nixon I, Saba NF, Hackenberg S, Specenier P, Worden F, Balsara B, Leoni M, Martell B, Scholz C, and Gualberto A
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, Female, Humans, Male, Middle Aged, Proto-Oncogene Mas, Quinolones adverse effects, Young Adult, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms genetics, Mutation, Missense, Proto-Oncogene Proteins p21(ras) genetics, Quinolones therapeutic use, Squamous Cell Carcinoma of Head and Neck drug therapy, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Purpose: Mutations in the HRAS (m HRAS ) proto-oncogene occur in 4%-8% of patients with recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC). Tipifarnib is a farnesyltransferase inhibitor that disrupts HRAS function. We evaluated the efficacy of tipifarnib in patients with R/M m HRAS HNSCC., Methods: We enrolled 30 patients with R/M HNSCC in a single-arm, open-label phase II trial of tipifarnib for m HRAS malignancies; one additional patient was treated on an expanded access program. After an ad hoc analysis of the first 16 patients with HNSCC with m HRAS variant allele frequency (VAF) data, enrollment was limited to those with a m HRAS VAF of ≥ 20% (high VAF). The primary end point was objective response rate. Secondary end points included assessing safety and tolerability. Patients received tipifarnib 600 or 900 mg orally twice daily on days 1-7 and 15-21 of 28-day cycles., Results: Of the 22 patients with HNSCC with high VAF, 20 were evaluable for response at the time of data cutoff. Objective response rate for evaluable patients with high-VAF HNSCC was 55% (95% CI, 31.5 to 76.9). Median progression-free survival on tipifarnib was 5.6 months (95% CI, 3.6 to 16.4) versus 3.6 months (95% CI, 1.3 to 5.2) on last prior therapy. Median overall survival was 15.4 months (95% CI, 7.0 to 29.7). The most frequent treatment-emergent adverse events among the 30 patients with HNSCC were anemia (37%) and lymphopenia (13%)., Conclusion: Tipifarnib demonstrated encouraging efficacy in patients with R/M HNSCC with HRAS mutations for whom limited therapeutic options exist (NCT02383927)., Competing Interests: Alan L. HoConsulting or Advisory Role: Bristol-Myers Squibb, Eisai, Genzyme, Merck, Novartis, Sun Pharma, Regeneron, TRM Oncology, Ayala Pharmaceuticals, AstraZeneca, Sanofi, CureVac, Prelude Therapeutics, Kura Oncology, McGivney Global Advisors, Rgenta, Exelixis, Genentech/Roche, AffyimmuneSpeakers' Bureau: Medscape, Omniprex America, NovartisResearch Funding: Lilly, Genentech/Roche, AstraZeneca, Bayer, Kura Oncology, Kolltan Pharmaceuticals, Eisai, Bristol-Myers Squibb, Astellas Pharma, Novartis, Merck, Pfizer, Ayala Pharmaceuticals, Allos Therapeutics, Daiichi Sankyo, Elevar TherapeuticsTravel, Accommodations, Expenses: Janssen Oncology, Merck, Kura Oncology, Ignyta, Ayala Pharmaceuticals, Klus Pharma Irene BranaConsulting or Advisory Role: Merck Sharp and Dohme, Rakuten Medical, Sanofi, Achilles Therapeutics, eTheRNA Immunotherapies, Cancer Expert NowSpeakers' Bureau: Bristol-Myers Squibb, Merck Serono, RocheResearch Funding: AstraZeneca, Bristol-Myers Squibb, Celgene, Gliknik, GlaxoSmithKline, Janssen Oncology, Kura Oncology, Merck Sharp and Dohme, Novartis, Orion Pharma GmbH, Pfizer, Roche, Shattuck Labs, Nanobiotix, Seattle GeneticsTravel, Accommodations, Expenses: AstraZeneca Spain, Merck Serono Robert HaddadEmployment: Dana-Farber Cancer InstituteLeadership: NCCNConsulting or Advisory Role: Celgene, Merck, Eisai, Bristol-Myers Squibb, AstraZeneca, Pfizer, Loxo, Genentech, Immunomic Therapeutics, GlaxoSmithKline, Gilead Sciences, Vaccinex, EMD Serono, BioNTech AG, Achilles TherapeuticsResearch Funding: Boehringer Ingelheim, Merck, Bristol-Myers Squibb, Celgene, AstraZeneca, VentiRx, Genentech, Pfizer, Kura OncologyPatents, Royalties, Other Intellectual Property: UpToDateOther Relationship: Nanobiotix, ISA Pharmaceuticals Jessica BaumanConsulting or Advisory Role: Pfizer, Bayer, AstraZeneca, Kura OncologyResearch Funding: Bristol-Myers SquibbTravel, Accommodations, Expenses: Trident Pharmaceuticals Sjoukje OostingResearch Funding: Celldex Deborah J. WongConsulting or Advisory Role: Bristol-Myers Squibb, Genentech/Roche, Sanofi/Aventis, Blueprint MedicinesResearch Funding: BioMed Valley Discoveries, Merck Serono, Merck Sharp and Dohme, ARMO BioSciences, AstraZeneca/MedImmune, Kura Oncology, Regeneron, Genentech/Roche, Bristol-Myers Squibb, FSTAR, Pfizer, Astellas Pharma, Enzychem Lifesciences, Lilly, Elevar Therapeutics Myung-Ju AhnHonoraria: AstraZeneca, Lilly, MSD, TAKEDAConsulting or Advisory Role: AstraZeneca, Boehringer Ingelheim, Lilly, MSD, TAKEDA, Alpha pharmaceutical Caroline EvenConsulting or Advisory Role: Innate Pharma, Bristol-Myers Squibb, MSD Oncology, Merck Serono Jerome FayetteHonoraria: AstraZeneca, Bristol-Myers Squibb, Merck Sharp and Dohme, Merck Serono, Innate Pharma, RocheConsulting or Advisory Role: AstraZeneca, Bristol-Myers Squibb, Merck Sharp and Dohme, Merck Serono, Innate Pharma, RocheResearch Funding: Bristol-Myers SquibbTravel, Accommodations, Expenses: Bristol-Myers Squibb, AstraZeneca, Merck Sharp and Dohme Kevin HarringtonHonoraria: Merck Sharp and Dohme, Amgen, Merck Serono, AstraZeneca/MedImmune, Boehringer Ingelheim, Bristol-Myers Squibb, Pfizer, Replimune, Oncolys BioPharma, MersanaConsulting or Advisory Role: Merck Sharp and Dohme, Merck Serono, AstraZeneca/MedImmune, Boehringer Ingelheim, Bristol-Myers Squibb, Pfizer, ReplimuneSpeakers' Bureau: Merck Sharp and Dohme, Merck Serono, Bristol-Myers SquibbResearch Funding: AstraZeneca, Merck Sharp and Dohme, Boehringer Ingelheim, Replimune Sung-Bae KimHonoraria: DAEHWA Pharmaceutical, ISU ABXISConsulting or Advisory Role: Lilly, AstraZeneca, DAEHWA Pharmaceutical, ISU AbxisResearch Funding: Novartis, Dongkook Pharma, Genzyme Lisa LicitraConsulting or Advisory Role: Eisai, Boehringer Ingelheim, AstraZeneca, SOBI, Novartis, Bayer, MSD, Merck Serono, Roche, Bristol-Myers Squibb, Incyte, Doxapharma, GlaxoSmithKline, Nanobiotix, Debiopharm Group, Amgen, IpsenResearch Funding: AstraZeneca, Novartis, Roche, MSD, Eisai, Merck Serono, Bristol-Myers Squibb, Boehringer Ingelheim, Celgene, Exelixis, IRX Therapeutics, Medpace, Pfizer, Debiopharm Group, RocheTravel, Accommodations, Expenses: Merck Serono, Bayer, Bristol-Myers Squibb, MSD, Eisai, AstraZeneca Nabil F. SabaHonoraria: Merck, Lilly, Pfizer, Bristol-Myers Squibb, CUE Biopharma, GlaxoSmithKline, Aduro Biotech, Kura Oncology, Genentech/RocheConsulting or Advisory Role: Pfizer, Bristol-Myers Squibb, Merck, Lilly, Bluprint, BiontechResearch Funding: Bristol-Myers Squibb, ExelixisTravel, Accommodations, Expenses: Bristol-Myers Squibb, Merck, Pfizer, Lilly, GlaxoSmithKline, Genentech/Roche, Bluprint Francis WordenHonoraria: Merck Sharp & Dohme, Eisai, Bristol-Myers Squibb, Bayer, RegeneronConsulting or Advisory Role: Merck, Loxo, Bristol-Myers Squibb, Eisai, Bayer, CUE Biopharma, Rakuten Medical, RegeneronResearch Funding: Pfizer, Merck, Eisai, Bristol-Myers Squibb, Loxo, Oragenics, LillyTravel, Accommodations, Expenses: Merck Sharp & Dohme, Bayer Binaifer BalsaraEmployment: Kura OncologyStock and Other Ownership Interests: Kura Oncology Mollie LeoniEmployment: Kura Oncology, Kyowa Kirin InternationalStock and Other Ownership Interests: Kura OncologyTravel, Accommodations, Expenses: Kura Oncology, Kyowa Kirin International Catherine ScholzEmployment: Kura Oncology, H3 BiomedicineStock and Other Ownership Interests: Kura OncologyPatents, Royalties, Other Intellectual Property: Methods of Treating Cancer Patients With Farnesyltransferase Inhibitors (FTI treatment of H-Ras mutant cancers), co-Inventor (014168-0011-999), Methods of Treating Cancer With Farnesyltransferase Inhibitors (FTI treatment of a CXCL12-expressing cancer), co-Inventor (014168-0021-999), Methods of Treating Cancer Patients With Farnesyltransferase Inhibitors (FTI treatment of Casitas B cell lymphoma [CBL] mutant cancers), co-Inventor (014168-0024-228), Therapies For Squamous Cell Carcinomas (FTI treatment of SCC with high frequencies of H-Ras mutant allele frequency), co-Inventor (014168-0051-888)Travel, Accommodations, Expenses: Kura OncologyNo other potential conflicts of interest were reported.
- Published
- 2021
- Full Text
- View/download PDF
15. Randomized, open-label phase 2 study comparing frontline dovitinib versus sorafenib in patients with advanced hepatocellular carcinoma.
- Author
-
Cheng AL, Thongprasert S, Lim HY, Sukeepaisarnjaroen W, Yang TS, Wu CC, Chao Y, Chan SL, Kudo M, Ikeda M, Kang YK, Pan H, Numata K, Han G, Balsara B, Zhang Y, Rodriguez AM, Zhang Y, Wang Y, and Poon RT
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Benzimidazoles adverse effects, Benzimidazoles pharmacokinetics, Biomarkers blood, Carcinoma, Hepatocellular blood, Carcinoma, Hepatocellular mortality, Cell Line, Tumor, Asia, Eastern epidemiology, Female, Humans, Liver Neoplasms blood, Liver Neoplasms mortality, Male, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Nude, Middle Aged, Niacinamide adverse effects, Niacinamide pharmacokinetics, Niacinamide therapeutic use, Phenylurea Compounds adverse effects, Phenylurea Compounds pharmacokinetics, Quinolones adverse effects, Quinolones pharmacokinetics, Sorafenib, Treatment Outcome, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Benzimidazoles therapeutic use, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy, Niacinamide analogs & derivatives, Phenylurea Compounds therapeutic use, Quinolones therapeutic use
- Abstract
Unlabelled: Angiogenesis inhibition by the vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR) inhibitor sorafenib provides survival benefit in hepatocellular carcinoma (HCC); however, angiogenic escape from sorafenib may occur due to angiogenesis-associated fibroblast growth factor receptor (FGFR) pathway activation. In addition to VEGFR and PDGFR, dovitinib inhibits FGFR. Frontline oral dovitinib (500 mg/day, 5 days on, 2 days off; n = 82) versus sorafenib (400 mg twice daily; n = 83) was evaluated in an open-label, randomized phase 2 study of Asian-Pacific patients with advanced HCC. The primary and key secondary endpoints were overall survival (OS) and time to tumor progression (TTP) as determined by a local investigator, respectively. Patients included in the study were ineligible for surgical and/or locoregional therapies or had disease progression after receiving these therapies. The median OS (95% confidence interval [CI]) was 8.0 (6.6-9.1) months for dovitinib and 8.4 (5.4-11.3) months for sorafenib. The median TTP (95% CI) per investigator assessment was 4.1 (2.8-4.2) months and 4.1 (2.8-4.3) months for dovitinib and sorafenib, respectively. Common any-cause adverse events included diarrhea (62%), decreased appetite (43%), nausea (41%), vomiting (41%), fatigue (35%), rash (34%), and pyrexia (30%) for dovitinib and palmar-plantar erythrodysesthesia syndrome (66%) and decreased appetite (31%) for sorafenib. Subgroup analysis revealed a significantly higher median OS for patients in the dovitinib arm who had baseline plasma soluble VEGFR1 (sVEGFR1) and hepatocyte growth factor (HGF) below median levels versus at or above the median levels (median OS [95% CI]: sVEGFR1, 11.2 [9.0-13.8] and 5.7 [4.3-7.0] months, respectively [P = .0002]; HGF, 11.2 [8.9-13.8] and 5.9 [5.0-7.6] months, respectively [P = 0.0002])., Conclusion: Dovitinib was well tolerated, but activity was not greater than sorafenib as a frontline systemic therapy for HCC. Based on these data, no subsequent phase 3 study has been planned. (Hepatology 2016;64:774-784)., (© 2016 by the American Association for the Study of Liver Diseases.)
- Published
- 2016
- Full Text
- View/download PDF
16. PDCD2 functions in cancer cell proliferation and predicts relapsed leukemia.
- Author
-
Barboza N, Minakhina S, Medina DJ, Balsara B, Greenwood S, Huzzy L, Rabson AB, Steward R, and Schaar DG
- Subjects
- Apoptosis Regulatory Proteins genetics, Biomarkers, Tumor genetics, Cell Line, Tumor, Gene Expression, Gene Knockdown Techniques, Hematopoietic Stem Cells metabolism, Humans, Leukemia, Myeloid, Acute pathology, Prospective Studies, RNA, Messenger genetics, RNA, Messenger metabolism, Apoptosis Regulatory Proteins metabolism, Biomarkers, Tumor metabolism, Cell Proliferation, Leukemia, Myeloid, Acute metabolism, Neoplasm Recurrence, Local metabolism
- Abstract
PDCD2 is an evolutionarily conserved eukaryotic protein with unknown function. The Drosophlia PDCD2 ortholog Zfrp8 has an essential function in fly hematopoiesis. Zfrp8 mutants exhibit marked lymph gland hyperplasia that results from increased proliferation of partially differentiated hemocytes, suggesting Zfrp8 may participate in cell growth. Based on the above observations we have focused on the role of PDCD2 in human cancer cell proliferation and hypothesized that aberrant PDCD2 expression may be characteristic of human malignancies. We report that PDCD2 is highly expressed in human acute leukemia cells as well as in normal hematopoietic progenitors. PDCD2 knockdown in cancer cells impairs their proliferation, but not viability relative to parental cells, supporting the notion that PDCD2 overexpression facilitates cancer cell growth. Prospective analysis of PDCD2 in acute leukemia patients indicates PDCD2 RNA expression correlates with disease status and is a significant predictor of clinical relapse. PDCD2's role in cell proliferation and its high expression in human malignancies make it an attractive, novel potential molecular target for new anti-cancer therapies.
- Published
- 2013
- Full Text
- View/download PDF
17. A pilot study of allogeneic cellular therapy for patients with advanced hematologic malignancies.
- Author
-
Medina DJ, Gharibo M, Savage P, Cohler A, Kuriyan M, Balsara B, Anand M, Schaar D, Krimmel T, Saggiomo K, Manago J, Talty L, Dudek L, Grospe S, Rubin A, and Strair RK
- Subjects
- Aged, Aged, 80 and over, Antigens, CD blood, Antigens, CD34 blood, Antineoplastic Combined Chemotherapy Protocols therapeutic use, CD3 Complex blood, CD8-Positive T-Lymphocytes immunology, Cell Transplantation, Female, Flow Cytometry, Hematologic Neoplasms immunology, Humans, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell surgery, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute surgery, Lymphoma, Non-Hodgkin immunology, Lymphoma, Non-Hodgkin surgery, Male, Middle Aged, Pilot Projects, Tissue Donors, Tissue Expansion methods, Tissue and Organ Harvesting methods, Transplantation, Homologous, Hematologic Neoplasms surgery
- Abstract
Allogeneic hematopoietic stem cell transplantation provides curative therapy for some patients with advanced hematologic malignancies. Disease response after allogeneic transplant is, at least in part, mediated by donor immune cells. In this report we describe a cellular therapy using haploidentical peripheral blood stem cells administered after very low dose total body irradiation (TBI) (100cGy). The donor cells were anticipated to be rejected, so no graft-versus-host (GVHD) prophylaxis was used. Patients with persistent disease beyond 8 weeks could be further treated with infusions of irradiated haploidentical donor cells. Of the 10 patients enrolled in the study, durable engraftment of allogeneic cells was seen in one patient. Two patients with resistant relapsed acute myelogenous leukemia (AML) had a disease response. Analysis of T cell reactivity from one patient who achieved a complete response but did not have durable engraftment of donor cells indicated that disease response was associated with the generation of host-derived anti-leukemic cytotoxic CD8+ T cells that reacted with an AML-associated proteinase 3 epitope. Results from this patient suggest that allogeneic therapy induced a host anti-tumor response associated with cytotoxic T cells reactive with a low affinity self-antigen.
- Published
- 2008
- Full Text
- View/download PDF
18. Nuclear factor-kappaB modulation in patients undergoing induction chemotherapy for acute myelogenous leukemia.
- Author
-
Strair RK, Gharibo M, Schaar D, Rubin A, Harrison J, Aisner J, Lin HC, Lin Y, Goodell L, Anand M, Balsara B, Dudek L, Rabson A, and Medina DJ
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cell Nucleus drug effects, Cell Nucleus metabolism, Choline administration & dosage, Enzyme-Linked Immunosorbent Assay, Female, Humans, Male, Middle Aged, NF-kappa B drug effects, Anti-Inflammatory Agents administration & dosage, Choline analogs & derivatives, Dexamethasone administration & dosage, Leukemia, Myeloid, Acute drug therapy, NF-kappa B biosynthesis, Salicylates administration & dosage
- Abstract
Purpose: Nuclear factor-kappaB (NF-kappaB) is constitutively expressed in many acute myelogenous leukemia (AML) cells and AML stem cells. Ex vivo treatment of AML cells with inhibitors of NF-kappaB results in diminished AML cell survival and enhances the cytotoxic effects of chemotherapeutic agents. The purpose of this study was to determine if standard anti-inflammatory agents modulate AML cell nuclear NF-kappaB when administered in conjunction with induction chemotherapy., Experimental Design: Patients with newly diagnosed AML were treated with dexamethasone, choline magnesium trisalicylate, or both for 24 hours prior to and 24 hours following initiation of standard induction chemotherapy. AML cell nuclear NF-kappaB was measured at baseline, 24, and 48 hours., Results: Choline magnesium trisalicylate +/- dexamethasone decreased nuclear NF-kappaB, whereas dexamethasone alone was associated with an increase in nuclear NF-kappaB in AML cells., Conclusions: These results show the feasibility of NF-kappaB modulation in conjunction with induction chemotherapy for patients with AML using inexpensive readily available medications. A follow-up study to determine the effects of NF-kappaB modulation on clinical end points is warranted.
- Published
- 2008
- Full Text
- View/download PDF
19. Comparative genomic hybridization of human breast epithelial cells transformed by estrogen and its metabolites.
- Author
-
Fernandez SV, Russo IH, Lareef M, Balsara B, and Russo J
- Subjects
- Breast cytology, Breast pathology, Dose-Response Relationship, Drug, Epithelial Cells, Female, Humans, Phenotype, Tumor Cells, Cultured, Aneuploidy, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Transformation, Neoplastic, DNA Damage, Estradiol pharmacology, Nucleic Acid Hybridization
- Abstract
The elevated incidence of breast cancer in women has been associated with prolonged exposure to high levels of estrogens. Our laboratory has demonstrated that treatment of the immortalized human breast epithelial cells MCF-10F with 17beta-estradiol (E2) or its metabolites 4-hydroxy-estradiol (4-OH-E2) and 2-hydroxy-estradiol (2-OH-E2) induces phenotypical changes indicative of neoplastic transformation. The MCF-10F cells treated with E2, 2-OH-E2 and 4-OH-E2 form colonies in agar methocel and lost their ductulogenic capacity in collagen matrix, expressing phenotypes similar to those induced by the carcinogen benz(a)pyrene (BP). To investigate whether these phenotypic changes were associated with genomic changes, MCF-10F cells treated with either E2, 2-OH-E2, or 4-OH-E2 at different doses (0.007 nM, 70 nM and 3.6 microM) were analyzed using a combination of standard G-banding and comparative genomic hybridization (CGH). Whereas no aneuploidy was observed in any of the transformed cells, the CGH revealed instead that only cells treated with 4-OH-E2 at the highest concentration (3.6 microM) exhibited DNA gains at 8q24, 9q34 and 20q13 and losses at 13q21.
- Published
- 2005
20. Chromosome 17p13.2 transfer reverts transformation phenotypes and Fas-mediated apoptosis in breast epithelial cells.
- Author
-
Lareef MH, Tahin Q, Song J, Russo IH, Mihaila D, Slater CM, Balsara B, Testa JR, Broccoli D, Grobelny JV, Mor G, Cuthbert A, and Russo J
- Subjects
- Animals, Benzo(a)pyrene toxicity, Breast enzymology, Breast pathology, Cell Division genetics, Cells, Cultured, Chromosomes, Human, Pair 11 genetics, Clone Cells cytology, Clone Cells enzymology, Collagen metabolism, Colony-Forming Units Assay, Epithelial Cells drug effects, Fibroblasts cytology, Fibroblasts enzymology, Humans, Hybrid Cells cytology, In Situ Hybridization, Fluorescence, Karyotyping, Mice, Microsatellite Repeats genetics, Phenotype, Polymerase Chain Reaction, Polymorphism, Restriction Fragment Length, Telomerase metabolism, Transfection, fas Receptor genetics, fas Receptor immunology, Apoptosis, Breast cytology, Cell Transformation, Neoplastic genetics, Chromosomes, Human, Pair 17 genetics, Epithelial Cells cytology, fas Receptor metabolism
- Abstract
Transformation of the human breast epithelial cells (HBEC) MCF-10F with the carcinogen benz(a)pyrene (BP) into BP1-E cells resulted in the loss of the chromosome 17 p13.2 locus (D17S796 marker) and formation of colonies in agar-methocel (colony efficiency (CE)), loss of ductulogenic capacity in collagen matrix, and resistance to anti-Fas monoclonal antibody (Mab)-induced apoptosis. For testing the role of that specific region of chromosome 17 in the expression of transformation phenotypes, we transferred chromosome 17 from mouse fibroblast donors to BP1-E cells. Chromosome 11 was used as negative control. After G418 selection, nine clones each were randomly selected from BP1-E-11neo and BP1-E-17neo hybrids, respectively, and tested for the presence of the donor chromosomes by fluorescent in situ hybridization and polymerase chain reaction-based restriction fragment length polymorphism (PCR-RFLP) analyses. Sensitivity to Fas Mab-induced apoptosis and evaluation of transformation phenotype expression were tested in MCF-10F, BP1-E, and nine BP1-E-11neo and BP1-E-17neo clones each. Six BP1-E-17neo clones exhibited a reversion of transformation phenotypes and a dose dependent sensitivity to Fas Mab-induced apoptosis, behaving similarly to MCF-10F cells. All BP1-E-11neo, and three BP1-E-17neo cell clones, like BP1-E cells, retained a high CE, loss of ductulogenic capacity, and were resistant to all Fas Mab doses tested. Genomic analysis revealed that those six BP1-E-17neo clones that were Fas-sensitive and reverted their transformed phenotypes had retained the 17p13.2 (D17S796 marker) region, whereas it was absent in all resistant clones, indicating that the expression of transformation phenotypes and the sensitivity of the cells to Fas-mediated apoptosis were under the control of genes located in this region., (Copyright 2004 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
21. Cloning and chromosomal localization of a gene encoding a novel serine/threonine kinase belonging to the subfamily of testis-specific kinases.
- Author
-
Visconti PE, Hao Z, Purdon MA, Stein P, Balsara BR, Testa JR, Herr JC, Moss SB, and Kopf GS
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Blotting, Northern, Chromosome Mapping, Chromosomes, Human, Pair 1 genetics, Cloning, Molecular, DNA, Complementary, Female, Gene Expression, Gene Library, Humans, In Situ Hybridization, Fluorescence, Male, Mice, Molecular Sequence Data, Organ Specificity, Protein Serine-Threonine Kinases chemistry, Reverse Transcriptase Polymerase Chain Reaction, Sequence Homology, Nucleic Acid, Protein Serine-Threonine Kinases genetics, Spermatozoa enzymology, Testis enzymology
- Abstract
Using reverse transcription-polymerase chain reaction (RT-PCR) with degenerate oligonucleotides corresponding to two highly conserved motifs within the protein kinase family of catalytic domains, we isolated a PCR fragment encoding a novel member of the testis-specific serine/threonine kinases (STK) from mouse male mixed germ cell mRNA. This PCR fragment recognized a 1020-bp transcript in male germ cells by northern blot analysis and was used to clone a full-length cDNA from a mouse mixed germ cell cDNA library. This cDNA has an open reading frame of 804 bases encoding a protein of 268 amino acids. This novel gene is almost identical to Stk22c, encoding a recently described testis-specific protein kinase, except for base-pair deletions that result in a shift in the coding region and an alteration of 22 amino acids (residues 109-131). Due to its homology with Stk22c, we have called this protein kinase gene Stk22d. Northern blot analysis revealed that this protein kinase is developmentally expressed in testicular germ cells and is not present in brain, ovary, kidney, liver, or early embryonic cells. We then cloned the human homologue of this protein kinase gene (STK22C) and found it to be expressed exclusively in the testis. Fluorescence in situ hybridization with both the human and mouse cDNA clones revealed syntenic localization on chromosomes 1p34-p35 and 4E1, respectively.
- Published
- 2001
- Full Text
- View/download PDF
22. Loss of heterozygosity analysis defines a 3-cM region of 15q commonly deleted in human malignant mesothelioma.
- Author
-
De Rienzo A, Balsara BR, Apostolou S, Jhanwar SC, and Testa JR
- Subjects
- Alleles, Chromosome Mapping, Chromosomes, Artificial, Yeast, Heterozygote, Humans, In Situ Hybridization, Fluorescence, Microsatellite Repeats, Models, Genetic, Polymorphism, Genetic, Tumor Cells, Cultured, Chromosome Deletion, Chromosomes, Human, Pair 15, Loss of Heterozygosity, Mesothelioma genetics
- Abstract
Previous comparative genomic hybridization and allelic loss analyses demonstrated frequent deletions from 15q11.1-15 in malignant mesothelioma. Recurrent losses of 15q11-22 have also been reported in several other tumor types such as breast and colorectal cancers. To more precisely map the commonly deleted region, we have performed a high density loss of heterozygosity analysis of 46 malignant mesotheliomas, using 26 polymorphic microsatellite markers spanning the entire long arm of chromosome 15. Allelic loss from 15q was observed in 22 of 46 (48%) cases. These analyses have defined a minimally deleted region of approximately 3-cM, which was confirmed to reside at 15q15 by fluorescence in situ hybridization analysis with yeast artificial chromosome probes. No tumor suppressor genes have been reported to map to this site. The minimally deleted region identified in this investigation overlaps those observed in other kinds of cancer, and is the smallest site of recurrent 15q loss identified to date in human tumors. The identification of this commonly deleted site implicates a putative tumor suppressor gene(s) at 15q15 involved in diverse forms of human neoplasia.
- Published
- 2001
- Full Text
- View/download PDF
23. Genomic imbalances in human lung adenocarcinomas and squamous cell carcinomas.
- Author
-
Pei J, Balsara BR, Li W, Litwin S, Gabrielson E, Feder M, Jen J, and Testa JR
- Subjects
- Chromosome Aberrations genetics, Chromosome Deletion, Chromosomes, Human, Pair 1 genetics, Chromosomes, Human, Pair 3 genetics, Chromosomes, Human, Pair 5 genetics, Chromosomes, Human, Pair 8 genetics, Gene Amplification genetics, Humans, Nucleic Acid Hybridization, Adenocarcinoma genetics, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Squamous Cell genetics, Lung Neoplasms genetics
- Abstract
Comparative genomic hybridization analysis was performed on 67 non-small-cell lung cancers (NSCLCs), including 32 squamous cell carcinomas (SCCs) and 35 adenocarcinomas (ACs), to identify differences in the patterns of genomic imbalance between these two histologic subtypes. Among the entire tumor set, the chromosome arms most often overrepresented were 1q, 3q, 5p, and 8q, each detected in 50-55% of cases. The most frequently underrepresented arms were 9q, 3p, 8p, and 17p. The number of imbalances was similar in SCCs and ACs (median number/case: 12 and 11, respectively). Moreover, many imbalances, such as gains of 1q, 5p, and 8q, occurred at a high frequency in both histologic subgroups. Several statistically significant differences, however, were found. The most prominent difference was gain of 3q24-qter, seen in 81% of SCCs compared with 31% of ACs (P < 0.0001), with amplification at 3q25-26 being detected in eight of 32 (25%) SCCs but in only two of 35 (6%) ACs. Gain of 20p13 and loss of 4q also were seen at a significantly higher rate in SCCs than in ACs, whereas overrepresentation of 6p was more common in ACs. Gains of 7q and 8q each were associated with higher-stage tumors and either positive nodal involvement or higher tumor grade. These data suggest that genes located in several chromosomal regions, particularly 3q25-26, may be associated with phenotypic properties that differentiate lung SCCs from ACs. Furthermore, certain imbalances, prominent among them gains of 7q and 8q, may be indicative of tumor aggressiveness in NSCLCs., (Copyright 2001 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
24. Human hepatocellular carcinoma is characterized by a highly consistent pattern of genomic imbalances, including frequent loss of 16q23.1-24.1.
- Author
-
Balsara BR, Pei J, De Rienzo A, Simon D, Tosolini A, Lu YY, Shen FM, Fan X, Lin WY, Buetow KH, London WT, and Testa JR
- Subjects
- Female, Gene Dosage, Humans, Male, Nucleic Acid Hybridization, Allelic Imbalance genetics, Carcinoma, Hepatocellular genetics, Chromosomes, Human, Pair 16 genetics, Liver Neoplasms genetics, Loss of Heterozygosity genetics
- Abstract
Comparative genomic hybridization (CGH) analysis was used to identify chromosomal imbalances in 52 human primary hepatocellular carcinomas (HCCs). The most prominent changes were gains of part or all of chromosome arms 8q (83% of cases) and 1q (73%) and loss of 16q (63%). Other commonly overrepresented sites were 5p, 7q, and Xq. Recurrent sites of DNA sequence amplification included 8q23--24 (five cases) and 11q13--14 (four cases). Other frequently underrepresented sites were 4q, 8p, 16p, and 17p. Taken collectively, these findings and data from other CGH studies of HCCs define a subset of chromosome segments that are consistently over- or underrepresented and highlight sites of putative oncogenes and tumor suppressor genes, respectively, involved in hepatocellular oncogenesis. Loss of heterozygosity analysis with a panel of polymorphic microsatellite markers distributed along 16q defined a minimal region of chromosomal loss at 16q23.1--24.1, suggesting that this region harbors a tumor suppressor gene whose loss/inactivation may contribute to the pathogenesis of many HCCs., (Copyright 2000 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
25. Identification and structural analysis of human RBM8A and RBM8B: two highly conserved RNA-binding motif proteins that interact with OVCA1, a candidate tumor suppressor.
- Author
-
Salicioni AM, Xi M, Vanderveer LA, Balsara B, Testa JR, Dunbrack RL Jr, and Godwin AK
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, COS Cells, Chromosome Mapping, Chromosomes, Human, Pair 14 genetics, Chromosomes, Human, Pair 5 genetics, Cloning, Molecular, Conserved Sequence, DNA, Complementary chemistry, DNA, Complementary genetics, DNA, Complementary isolation & purification, Female, Gene Expression, Genes, Tumor Suppressor, Humans, In Situ Hybridization, Fluorescence, Male, Microscopy, Fluorescence, Minor Histocompatibility Antigens, Models, Molecular, Molecular Sequence Data, Protein Binding, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Structure, Tertiary, Proteins genetics, Proteins metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, RNA-Binding Proteins chemistry, RNA-Binding Proteins metabolism, Sequence Alignment, Sequence Analysis, DNA, Sequence Homology, Amino Acid, Tissue Distribution, RNA-Binding Proteins genetics, Tumor Suppressor Proteins
- Abstract
The OVCA1 gene is a candidate for the breast and ovarian tumor suppressor gene at chromosome 17p13.3. To help determine the function(s) of OVCA1, we used a yeast two-hybrid screening approach to identify OVCA1-associating proteins. One such protein, which we initially referred to as BOV-1 (binder of OVCA1-1) is 173 or 174 amino acids in length and appears to be a new member of a highly conserved RNA-binding motif (RBM) protein family that is highly conserved evolutionarily. Northern blot analysis revealed that BOV-1 is ubiquitously expressed and that three distinct messenger RNA species are expressed, 1-, 3.2-, and 5.8-kb transcripts. The 1-kb transcript is the most abundant and is expressed at high levels in the testis, heart, placenta, spleen, thymus, and lymphocytes. Using fluorescence in situ hybridization and the 5.8-kb complementary DNA probe, we determined that BOV-1 maps to both chromosome 5q13-q14 and chromosome 14q22-q23. Further sequence analysis determined that the gene coding the 1- and the 3.2-kb transcripts (HGMW-approved gene symbol RBM8A) maps to 14q22-q23, whereas a second highly related gene coding for the 5.8-kb transcript resides at chromosome 5q13-q14 (HGMW-approved gene symbol RBM8B). The predicted proteins encoded by RBM8A and RBM8B are identical except that RBM8B is 16 amino acids shorter at its N-terminus. Molecular modeling of the RNA-binding domain of RBM8A and RBM8B, based on homology to the sex-lethal protein of Drosophila, identifies conserved residues in the RBM8 protein family that are likely to contact RNA in a protein-RNA complex. The conservation of sequence and structure through such an evolutionarily divergent group of organisms suggests an important function for the RBM8 family of proteins., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
26. Novel human and mouse annexin A10 are linked to the genome duplications during early chordate evolution.
- Author
-
Morgan RO, Jenkins NA, Gilbert DJ, Copeland NG, Balsara BR, Testa JR, and Fernandez MP
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Chromosome Mapping, Chromosomes genetics, Chromosomes, Human, Pair 4 genetics, Crosses, Genetic, DNA, Complementary chemistry, DNA, Complementary genetics, Female, Humans, In Situ Hybridization, Fluorescence, Male, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Muridae, Phylogeny, Sequence Alignment, Sequence Analysis, DNA, Sequence Homology, Amino Acid, Sequence Homology, Nucleic Acid, Annexins genetics, Chordata, Nonvertebrate genetics, Evolution, Molecular, Gene Duplication
- Abstract
We have identified and characterized a 12th subfamily of vertebrate annexins by systematic analysis of the primary structure, chromosomal mapping, and molecular evolution of unique cDNA and protein sequences from human and mouse. Distinctive features included rare expression, a codon deletion in conserved repeat 3, and an unusual ablation of the type II calcium-binding sites in tetrad core repeats 1, 3, and 4. The paralogy of novel annexin A10 (following revised nomenclature) was confirmed by FISH-mapping human ANXA10 to chromosome 4q33 and genetic linkage mapping mouse Anxa10 to midchromosome 8. Phylogenetic analysis established that the 5' and 3' halves of the annexin A6 octad are more closely related to annexins A5 and A10, respectively, than they are to each other. Molecular date estimates, paralogy linkage maps between human chromosomes 4 and 5, and annexin structural considerations led to the proposal that annexins A5 and A10 may have been the direct progenitors of annexin A6 octad formation via chromosomal duplication during the genome expansion in early chordates., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
27. Comparative genomic hybridization and loss of heterozygosity analyses identify a common region of deletion at 15q11.1-15 in human malignant mesothelioma.
- Author
-
Balsara BR, Bell DW, Sonoda G, De Rienzo A, du Manoir S, Jhanwar SC, and Testa JR
- Subjects
- DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Humans, Nucleic Acid Hybridization, Rad51 Recombinase, Tumor Cells, Cultured, Chromosomes, Human, Pair 15, Loss of Heterozygosity, Mesothelioma genetics
- Abstract
Comparative genomic hybridization analysis was performed to identify chromosomal imbalances in 24 human malignant mesothelioma (MM) cell lines derived from untreated primary tumors. Chromosomal losses accounted for the majority of genomic imbalances. The most frequent underrepresented segments were 22q (58%) and 15q1.1-21 (54%); other recurrent sites of chromosomal loss included 1p12-22 (42%), 13q12-14 (42%), 14q24-qter (42%), 6q25-qter (38%), and 9p21 (38%). The most commonly overrepresented segment was 5p (54%). DNA sequence amplification at 3p12-13 was observed in two cases. Whereas some of the regions of copy number decreases (i.e., segments in 1p, 6q, 9p, and 22q) have previously been shown to be common sites of karyotypic and allelic loss in MM, our comparative genomic hybridization analyses identified a new recurrent site of chromosomal loss within 15q in this malignancy. To more precisely map the region of 15q deletion, loss of heterozygosity analyses were performed with a panel of polymorphic microsatellite markers distributed along 15q, which defined a minimal region of chromosomal loss at 15q11.1-15. The identification of frequent losses of a discrete segment in 15q suggests that this region harbors a putative tumor suppressor gene whose loss/inactivation may contribute to the pathogenesis of many MMs.
- Published
- 1999
28. Characterization of MOAT-C and MOAT-D, new members of the MRP/cMOAT subfamily of transporter proteins.
- Author
-
Belinsky MG, Bain LJ, Balsara BB, Testa JR, and Kruh GD
- Subjects
- ATP-Binding Cassette Transporters genetics, Amino Acid Sequence, Anion Transport Proteins, Carrier Proteins genetics, Chromosomes, Human, Pair 17 genetics, Chromosomes, Human, Pair 3 genetics, DNA, Neoplasm isolation & purification, Female, Gene Expression Regulation, Neoplastic, Humans, Molecular Sequence Data, Multidrug Resistance-Associated Proteins, Neoplasm Proteins genetics, Sequence Analysis, DNA, ATP-Binding Cassette Transporters chemistry, Carrier Proteins chemistry, DNA, Neoplasm analysis, Leukemia genetics, Neoplasm Proteins chemistry, Ovarian Neoplasms genetics
- Abstract
Background: Multidrug resistance-associated protein (MRP) and canalicular multispecific organic anion transporter (cMOAT) are transporter proteins that pump organic anions across cellular membranes and have been linked to resistance to cytotoxic drugs. We previously identified MOAT-B, an MRP/cMOAT-related transporter, by use of a polymerase chain reaction approach. However, analysis of expressed sequence tag (EST) databases indicated that there might be additional MRP/cMOAT-related transporters. To further define the MRP/cMOAT subfamily of transporters, we used EST probes to isolate complementary DNAs for two related transporter proteins, MOAT-C and MOAT-D., Methods: MOAT-C and MOAT-D expression patterns in human tissues were determined by RNA blot analysis, and chromosomal localization of the genes was determined by fluorescence in situ hybridization., Results: MOAT-C is predicted to encode a 1437-amino-acid protein that, among eukaryotic transporters, is most closely related to MRP, cMOAT, and MOAT-B (about 36% identity). However, MOAT-C is less related to MRP and cMOAT than MRP and cMOAT are to each other (about 48% identity). Like MOAT-B, MOAT-C lacks an N-terminal membrane-spanning domain, indicating that the topology of this protein is similarly distinct from that of MRP and cMOAT. MOAT-D is predicted to encode a 1527-amino-acid protein that is the closest known relative of MRP (about 58% identity). MOAT-D is also highly related to cMOAT (about 47% identity). The presence of an N-terminal membrane-spanning domain indicates that the topology of MOAT-D is quite similar to that of MRP and cMOAT. MOAT-C transcripts are widely expressed in human tissues; however, MOAT-D transcript expression is more restricted. The MOAT-C and MOAT-D genes are located at chromosomes 3q27 and 17q21.3, respectively., Conclusions: On the basis of amino acid identity and protein topology, the MRP/cMOAT transporter subfamily falls into two groups; the first group consists of MRP, cMOAT, and MOAT-D, and the second group consists of MOAT-B and MOAT-C.
- Published
- 1998
- Full Text
- View/download PDF
29. Combined chromosome microdissection and comparative genomic hybridization detect multiple sites of amplification DNA in a human lung carcinoma cell line.
- Author
-
Taguchi T, Cheng GZ, Bell DW, Balsara B, Liu Z, Siegfried JM, and Testa JR
- Subjects
- Blotting, Southern, Chromosome Banding, Chromosomes, Human, Pair 7 genetics, Chromosomes, Human, Pair 8 genetics, Chromosomes, Human, Pair 9 genetics, DNA, Neoplasm analysis, Epidermal Growth Factor genetics, Genes, myc genetics, Humans, In Situ Hybridization, Fluorescence, Polymerase Chain Reaction, Tumor Cells, Cultured, Carcinoma, Non-Small-Cell Lung genetics, Chromosome Mapping, Chromosomes, Human, Pair 10 genetics, Gene Amplification genetics, Lung Neoplasms genetics
- Abstract
Chromosome microdissection-fluorescence in situ hybridization and comparative genomic hybridization (CGH) were performed in parallel to identify the native location of amplified DNA in a human non-small cell lung cancer (NSCLC) cell line exhibiting a homogeneously staining region (hsr) and double minutes (dmin). The native locations of microdissected DNA from the hsr and dmin were 7p12-13 and 8q24, respectively. Southern analysis revealed coamplification of EGFR (7p12) and MYC (8q24). CGH detected amplification of DNA not only from 7p12-13 and 8q24, but also from 9p24 and 10q22.
- Published
- 1997
- Full Text
- View/download PDF
30. Characterization of unconventional MYO6, the human homologue of the gene responsible for deafness in Snell's waltzer mice.
- Author
-
Avraham KB, Hasson T, Sobe T, Balsara B, Testa JR, Skvorak AB, Morton CC, Copeland NG, and Jenkins NA
- Subjects
- Amino Acid Sequence, Animals, Female, Humans, Mice, Molecular Sequence Data, Myosin Heavy Chains biosynthesis, Chromosomes, Human, Pair 6, Deafness genetics, Myosin Heavy Chains genetics
- Abstract
Deafness is the most common form of sensory impairment in humans. Mutations in unconventional myosins have been found to cause deafness in humans and mice. The mouse recessive deafness mutation, Snell's waltzer, contains an intragenic deletion in an unconventional myosin, myosin VI (locus designation, Myo6). The requirement for Myo6 for proper hearing in mice makes this gene an excellent candidate for a human deafness disorder. Here we report the cloning and characterization of the human unconventional myosin VI (locus designation, MYO6) cDNA. The MYO6 gene maps to human chromosome 6q13. The isolation of the human gene makes it now possible to determine if mutations in MYO6 contribute to the pathogenesis of deafness in the human population.
- Published
- 1997
- Full Text
- View/download PDF
31. Comparative genomic hybridization analysis detects frequent, often high-level, overrepresentation of DNA sequences at 3q, 5p, 7p, and 8q in human non-small cell lung carcinomas.
- Author
-
Balsara BR, Sonoda G, du Manoir S, Siegfried JM, Gabrielson E, and Testa JR
- Subjects
- Biopsy, Carcinoma, Non-Small-Cell Lung metabolism, Chromosomes, Human, Pair 3, Chromosomes, Human, Pair 5, Chromosomes, Human, Pair 7, Chromosomes, Human, Pair 8, Female, Humans, Lung Neoplasms metabolism, Male, Nucleic Acid Hybridization, Sequence Analysis, DNA, Tumor Cells, Cultured, Carcinoma, Non-Small-Cell Lung genetics, DNA, Neoplasm metabolism, Gene Amplification, Lung Neoplasms genetics
- Abstract
Comparative genomic hybridization analysis was used to identify chromosomal imbalances in 20 non-small cell lung carcinoma (NSCLC) biopsies and cell lines. The chromosome arms most often overrepresented were 3q (85%), 5p (70%), 7p (65%), and 8q (65%), which were observed at high copy numbers in many cases. Other common overrepresented sites were 1q, 2p, and 20p. DNA sequence amplification was often observed, with the most frequent site being 3q26 (six cases). Other recurrent sites of amplification included 8q24, 3q13, 3q28-qter, 7q11.2, 8p11-12, 12p12, and 19q13.1-13.2. The most frequent underrepresented segment was 3p21 (50%); other recurrent sites of autosomal loss included 8p21-pter, 15q11.2-13, 5q11.2-15, 9p, 13q12-14, 17p, and 18q21-qter. These regions of copy number decreases are also common sites of allelic loss, further implicating these sites as locations of tumor suppressor genes. Although some of the overrepresented segments harbor known or suspected oncogenes/growth-regulatory genes, we have identified 3q and 5p as new sites that are very frequently overrepresented in NSCLC. These findings could represent entry points for the identification of novel amplified DNA sequences that may contribute to the development or progression of NSCLC.
- Published
- 1997
32. Loss of heterozygosity analysis defines a critical region in chromosome 1p22 commonly deleted in human malignant mesothelioma.
- Author
-
Lee WC, Balsara B, Liu Z, Jhanwar SC, and Testa JR
- Subjects
- Chromosomes, Human, Pair 1 ultrastructure, Humans, In Situ Hybridization, Polymerase Chain Reaction, Polymorphism, Genetic, Saccharomyces cerevisiae genetics, Chromosomes, Human, Pair 1 genetics, Mesothelioma genetics, Sequence Deletion
- Abstract
Previous cytogenetic analysis has revealed frequent losses of chromosome 1p21-22 in human malignant mesothelioma, suggesting that the loss or inactivation of a tumor suppressor gene's) residing at this site may contribute to the tumorigenic conversion of mesothelial cells. To more precisely define the location of the target gene, primary tumor specimens and cell lines from 50 malignant mesotheliomas were examined for loss of heterozygosity using short tandem repeat polymorphism (STRP) markers. Nineteen STRP markers established by the Cooperative Human Linkage Center were selected for the initial screening of the entire short arm of chromosome 1. Thirty-seven cases (74%) showed allelic losses at least at one locus in 1p. Thirty-six of these cases showed losses of 1p21-22, including 23 with partial deletions involving this region. To obtain a higher resolution map of this region, another 13 STRP markers from the Genethon map were used to define the shortest region of overlapping deletions to a 4-cM segment flanked by the loci D1S435 and D1S236. The chromosomal location of the critically deleted region was confirmed to be within 1p22 by karyotypic and fluorescence in situ hybridization analyses.
- Published
- 1996
33. Cytogenetic studies on a patient with prepubertal breast cancer: a case report.
- Author
-
Balsara BR, Varughese T, Bhat AV, Rao RS, and Bhisey AN
- Subjects
- Breast Neoplasms therapy, Child, Female, Humans, Breast Neoplasms genetics, Chromosome Aberrations
- Abstract
Cytogenetic studies were carried in a 10 year old girl with prepubertal breast cancer for assessing inherited genetic susceptibility to chromosome breakage. The girl presented with a tumour in the left breast. Histologically it was diagnosed as secretory carcinoma (SC). Chromosome anomalies observed in phytohemagglutinin (PHA-P) stimulated lymphocytes were del(2)(q33), del(3)(p24), del(7)(q22) and dup(12)(p11p12). The regions involved have been reported in breast tumors. These loci, detected in peripheral blood lymphocytes (PBL), could be the sites susceptible to breakage, its subsequent effect being manifested in the target (breast) tissue.
- Published
- 1996
- Full Text
- View/download PDF
34. Cytogenetic studies on patients of acute lymphoblastic leukemia Burkitt's type with (8;14) & (14;18) translocations.
- Author
-
Gladstone B, Kadam PR, Balsara BR, Pai SK, Gopal R, Nair CN, Parikh PM, Saikia T, and Advani SH
- Subjects
- Adult, Child, Child, Preschool, Female, Humans, Male, Burkitt Lymphoma genetics, Chromosomes, Human, Pair 14, Chromosomes, Human, Pair 18, Chromosomes, Human, Pair 8, Translocation, Genetic
- Abstract
Eight patients with acute lymphoblastic leukemia of Burkitt's type (ALL-L3) and two patients with Burkitt's lymphoma (BL) were subjected for cytogenetic studies. Translocation (8;14)(q24;q32) was present in nine (90%) patients; seven patients of ALL-L3 and two of BL. One ALL-L3 patient revealed t(14;18)(q32;q21) in 100 per cent metaphases. Additional clonal chromosomal anomalies present in these patients were deletion (6q) (40%) and trisomy 21(20%). The occurrence of t(8;14)(q24;q32) in ALL-L3 and BL patients in our series supports the association of t(8;14) with ALL-L3 and Burkitt's lymphoma.
- Published
- 1994
35. Flow cytometric DNA analysis of squamous cell carcinomas of the oral cavity: correlation with clinical and histopathological features.
- Author
-
Balsara BR, Borges AM, Pradhan SA, Rajpal RM, and Bhisey AN
- Subjects
- Adult, Aged, Carcinoma, Squamous Cell pathology, Female, Flow Cytometry, Humans, Lymphatic Metastasis, Male, Middle Aged, Mouth Neoplasms pathology, Neck, S Phase, Carcinoma, Squamous Cell genetics, DNA, Neoplasm analysis, Mouth Neoplasms genetics, Ploidies
- Abstract
DNA content was measured in 68 squamous cell carcinomas (SCC) of the oral cavity by flow cytometry. Samples fixed in 95% alcohol and disaggregated with 0.5% pepsin were stained with 4,6-diamidino-phenylindole (DAPI) for flow cytometry. The tumours were classified according to the TNM classification--1987, and graded histopathologically. A positive correlation between tumour size and ploidy status was observed. Poorly differentiated tumours were mainly non-diploid (P < 0.01, chi 2). A majority of the node positive (N+) tumours were non-diploid (P < 0.05). It was possible to distinguish diploid, N+, T4 tumours from diploid, N-, T4 tumours by their higher S-phase fraction (SpF). SpF was also a useful parameter to differentiate diploid, N+ tumours from diploid, N- tumours among the moderately differentiated (MD) SCC. These results suggest that ploidy and SpF can be useful correlates of tumour behaviour.
- Published
- 1994
- Full Text
- View/download PDF
36. DNA analysis of breast cancer by flow cytometry & correlation with other prognostic parameters.
- Author
-
Redkar AA, Balsara BR, Bhisey AN, Sampat MB, and Mittra I
- Subjects
- Breast Neoplasms pathology, Female, Humans, India, S Phase, Aneuploidy, Breast Neoplasms genetics, DNA, Neoplasm analysis, Diploidy, Flow Cytometry
- Abstract
Flow cytometric estimation of DNA content (ploidy and S-phase fraction--SpF) was done on breast cancer tissues from 171 patients. Twenty eight per cent of the tumours were diploid and 72 per cent were aneuploid. SpF was measurable in 82 DNA histograms; of these 22.4 per cent had SpF less than 10 per cent, 34.1 per cent had SpF between 10-20 and 43.5 per cent had SpF greater than 20 per cent. The mean SpF of the measurable histograms was 19.01 per cent with a range 1.78 to 45.19 per cent. A significant correlation between DNA ploidy and SpF was observed (P less than 0.01). Eighty nine per cent of diploid tumours had SpF less than 10 per cent and 73 per cent of aneuploid tumours had SpF greater than 20 per cent. A significant correlation was also found between ploidy and SpF and oestrogen receptor (ER) status of the tumours (P less than 0.05) and between SpF and progesterone receptor (PgR) status of the tumours (P less than 0.05), but not between ploidy and PgR status of the tumours. A significant direct correlation was observed between SpF and tumour grade (P less than 0.05), but not between ploidy and tumour grade. No correlation was observed between DNA ploidy and SpF and tumour type, tumour size, axillary lymph node involvement, age and menopausal status of the patients. Although the incidence of breast cancer is one-third of that reported in the Western countries, there is apparently no biological difference between the various parameters studied.
- Published
- 1992
37. Cytogenetic features of erythroleukemia (EL). A study of 11 cases.
- Author
-
Kadam PR, Balsara BR, Zafaraullah KZ, Dadabhoy KD, Bhisey AN, and Advani SH
- Subjects
- Adolescent, Adult, Blood Cell Count, Child, Child, Preschool, Female, Gene Rearrangement, Humans, Infant, Karyotyping, Male, Middle Aged, Chromosome Aberrations, Leukemia, Erythroblastic, Acute genetics
- Abstract
Cytogenetic studies were carried on 11 patients with erythroleukemia (EL). Most of these patients showed major chromosomal abnormalities (MAKA), karyotypic instability, and complex chromosomal rearrangements. On the basis of the cytogenetic criteria, 10 patients could be distinguished into erythroid (9 cases) and myeloid types of EL (1 case). The patients did not show any consistent chromosomal abnormality. However, abnormalities of chromosomes 1, 3, 7, 8, 16, and 17 were seen in more than one patient. In patients with the erythroid type of EL, besides the MAKA pattern, three patients showed increased frequency of hyperdiploid polyploid cells ranging from triploidy to tetraploidy.
- Published
- 1990
- Full Text
- View/download PDF
38. Establishment and characterization of four new squamous cell carcinoma cell lines derived from oral tumors.
- Author
-
Tatake RJ, Rajaram N, Damle RN, Balsara B, Bhisey AN, and Gangal SG
- Subjects
- Carcinoma, Squamous Cell analysis, Carcinoma, Squamous Cell ultrastructure, DNA, Neoplasm analysis, Flow Cytometry, Histocompatibility Antigens Class I analysis, Histocompatibility Antigens Class II analysis, Humans, Keratins analysis, Mouth Neoplasms analysis, Mouth Neoplasms ultrastructure, Tumor Cells, Cultured, Carcinoma, Squamous Cell pathology, Mouth Neoplasms pathology
- Abstract
Four cell lines were established from squamous cell carcinomas (SCC) of the oral cavity. Cell lines AW 13516 and AW 8507 were derived from poorly differentiated SCC and epidermoid carcinoma of the tongue respectively. Cell line AW 10498 was derived from moderately differentiated SCC of the lower alveolus, and AW 9803 grew from a well-differentiated SCC of a retromolar trigone. The cultures showed typical epithelial cell morphology, numerous mitotic figures, occasional multinucleated giant cells, individual cell diskeratosis and nuclear and nucleolar abnormalities. The cell lines AW 13516 and AW 8507 were fast growers with a doubling time of 35.5 h and 31.9 h, respectively, which was independent of the initial seeding density. Cell lines AW 10498 (doubling time 52.2 h) and AW 9803 (doubling time 66 h) showed slower growth and had shorter doubling times at higher seeding densities. The presence of cytokeratins was detected in all the four cell lines by using polyclonal antikeratin antisera in indirect immunofluorescence and in Western blotting. None of the cell lines expressed major histocompatibility complex (MHC) class II antigens. MHC class I antigens were expressed by three cell lines but not by AW 9803. Flow cytometric analysis of DNA content and chromosomal studies suggested the presence of polyploidy and aneuploidy in all the four cell lines. Ultrastructural studies revealed typical epithelial cell features, such as the presence of desmosomes, tonofilaments and keratin bundles.
- Published
- 1990
- Full Text
- View/download PDF
39. Immunological, mutagenic & genotoxic investigations in gas exposed population of Bhopal.
- Author
-
Deo MG, Gangal S, Bhisey AN, Somasundaram R, Balsara B, Gulwani B, Darbari BS, Bhide S, and Maru GB
- Subjects
- Adolescent, Adult, Antibody Formation drug effects, Child, Chromosome Aberrations drug effects, Cyanates poisoning, Female, Humans, Immunity, Cellular drug effects, India, Male, Middle Aged, Accidents, Occupational, Disasters, Gas Poisoning etiology, Gas Poisoning genetics, Gas Poisoning immunology, Isocyanates
- Published
- 1987
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.