151
|
Massard C, Voigt JJ, Laplanche A, Culine S, Lortholary A, Bugat R, Theodore C, Priou F, Kaminsky MC, Lesimple T, Pivot X, Coudert B, Douillard JY, Merrouche Y, Fizazi K. Carcinoma of an unknown primary: are EGF receptor, Her-2/neu, and c-Kit tyrosine kinases potential targets for therapy? Br J Cancer 2007; 97:857-61. [PMID: 17876336 PMCID: PMC2360401 DOI: 10.1038/sj.bjc.6603942] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Carcinomas of an unknown primary site (CUP) are heterogeneous tumours with a median survival of only 8 months. Tyrosine kinase inhibitors are promising new drugs. The aim of this study was to determine the expression of EGF-receptor, Her-2/neu, and c-Kit tyrosine kinases in CUP. Paraffin-embedded specimens were obtained from 54 patients with a CUP who were included in the GEFCAPI 01 randomised phase II trial. Immunohistochemistry was performed using the Dako autostainer with antibodies directed against HER-2/neu protein, EGFR protein, and c-Kit protein (CD117). EGFR expression was found in 36 out of 54 samples (66%). In contrast, Her-2/neu overexpression and c-Kit positivity were only detected in 4 and 10% of patients, respectively. No significant association was found between the expression of the tyrosine kinase receptors and prognosis. EGFR expression was significantly associated with response to cisplatin-based chemotherapy: the response rates were 50 and 22% in patients with EGFR-positive tumours and EGFR-negative tumours, respectively (P<0.05). This study shows that EGFR is frequently expressed in CUP. This finding may prompt clinical trials investigating EGFR inhibitors in this setting. In contrast, c-Kit expression and Her-2/neu overexpression occur infrequently in CUP. EGFR expression was correlated to tumour chemosensitivity.
Collapse
Affiliation(s)
- C Massard
- Department of Medicine, Institut Gustave Roussy, 39 rue Camille Desmoulins, Villejuif 94805, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Aoki M, Nabeshima K, Koga K, Hamasaki M, Suzumiya J, Tamura K, Iwasaki H. Imatinib mesylate inhibits cell invasion of malignant peripheral nerve sheath tumor induced by platelet-derived growth factor-BB. J Transl Med 2007; 87:767-79. [PMID: 17558420 DOI: 10.1038/labinvest.3700591] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is rare, highly aggressive, resistant to radiochemotherapy, and associated with poor prognosis. Basic research to develop new treatment regimes is critically needed. This study was designed to identify motogenic factor(s) involved in MPNST cell invasion and inhibitor(s) of such invasive activity. We profiled the invasion-inducing activities of eight motogenic growth factors on two human MPNST cell lines, FU-SFT8611 and 9817, using in vitro Matrigel invasion assays. Platelet-derived growth factor-BB (PDGF-BB) was identified as the most effective MPNST cell invasion-inducing factor. Epidermal growth factor (EGF) and hepatocyte growth factor (HGF) also stimulated invasion in one MPNST cell line. Expressions of PDGF-BB and EGF receptors (PDGFR-beta and EGFR) mRNAs were detected more frequently and their proteins were expressed at higher levels in MPNST tissues than benign peripheral nerve sheath tumors (schwannomas and neurofibromas). In both MPNST cell lines, PDGF-BB induced tyrosine phosphorylation of PDGFR-beta but not of PDGFR-alpha, and specific PDGFR-beta inhibition by small interfering RNA to the receptor inhibited PDGF-BB-stimulated MPNST cell invasion, suggesting the predominant role of PDGFR-beta. Inhibition of PDGFR-beta phosphorylation by pretreatment with herbimycin A and imatinib mesylate effectively suppressed basement membrane invasion and cell growth in vitro. No mutations were present in exons 12 and 18 of PDGFR-beta in both MPNST cell lines and 10 human MPNST tissues examined. Our results indicated that PDGF-BB enhanced the invasive activity of MPNST cells through PDGFR phosphorylation and that imatinib inhibited such activity. The results provide the ground for further assessment of the therapeutic potential of imatinib in suppressing the invasion and growth of MPNST.
Collapse
Affiliation(s)
- Mikiko Aoki
- Department of Pathology, Fukuoka University School of Medicine, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
153
|
New approaches to the management of Philadelphia-chromosome-positive acute lymphocytic leukemia. Curr Hematol Malig Rep 2007; 2:183-9. [PMID: 20425368 DOI: 10.1007/s11899-007-0025-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The discovery of targeted ABL tyrosine kinase inhibitors has allowed significant advances in the treatment of de novo Philadelphia chromosome (Ph)-positive ALL. Whereas the outcome with standard chemotherapy was previously dismal, the use of imatinib in front-line therapy has improved relapse-free survival and overall survival, even in the absence of allogeneic stem cell transplantation in first complete remission (particularly for those with comorbidities or lack of a suitable donor). However, the emergence of resistance to imatinib presents a new therapeutic challenge. Novel tyrosine kinase inhibitors with enhanced inhibitory potency against ABL and other kinases may further improve on the results observed with imatinib. Optimal use of these novel agents in the treatment schema of Ph-positive ALL will be paramount in ensuring continued success in the eradication of this disease.
Collapse
|
154
|
Preto A, Moutinho C, Velho S, Oliveira C, Rebocho AP, Figueiredo J, Soares P, Lopes JM, Seruca R. A subset of colorectal carcinomas express c-KIT protein independently of BRAF and/or KRAS activation. Virchows Arch 2007; 450:619-26. [PMID: 17487504 DOI: 10.1007/s00428-007-0420-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Revised: 03/28/2007] [Accepted: 04/05/2007] [Indexed: 10/23/2022]
Abstract
c-KIT is a tyrosine kinase receptor found to be overexpressed in several tumours, namely, GISTs, breast, lung, prostate, ovarian and colorectal carcinomas (CRC). We aimed at determining the frequency of c-KIT expression and mutations in a series of 109 CRC cases (73 primary tumours and 36 lymph node metastases) characterised for KRAS and BRAF mutations. We also aimed at analysing the cellular effects of STI571/Gleevec in CRC-derived cell lines displaying c-KIT expression and KRAS or BRAF mutations. By immunohistochemistry, we found c-KIT overexpression in 15% (11/73) of primary tumours and in 14% (5/36) of metastasis; however, cases showing overexpression did not show c-kit mutations in hotspot regions. The majority (64%) of primary tumours with c-KIT overexpression had mutations at KRAS-BRAF genes. The same was true for 60% of the metastases. We treated CRC cell lines with STI571/Gleevec and verified that it inhibits proliferation and induces apoptosis in all cell lines. In conclusion, overexpression of c-KIT is observed in a subset of primary and CRC metastases in the absence of c-kit mutations. STI571/Gleevec increases apoptosis in CRC cell lines independently of its genetic profile, suggesting that STI571/Gleevec is likely to be an alternative drug for the clinical trials of CRC.
Collapse
Affiliation(s)
- Ana Preto
- Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr Roberto Frias s/n, 4200-465 Porto, Portugal.
| | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Hong SM, Hwang I, Song DE, Choi J, Yu E. Clinical and prognostic significances of nuclear and cytoplasmic KIT expressions in extrahepatic bile duct carcinomas. Mod Pathol 2007; 20:562-9. [PMID: 17396144 DOI: 10.1038/modpathol.3800771] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
After receiving FDA approval as a therapeutic regimen in gastrointestinal stromal tumors, the tyrosine kinase inhibitor imatinib mesylate has been applied to the treatment of other solid malignant neoplasms. To evaluate the usefulness of imatinib mesylate as a possible therapeutic regimen in extrahepatic bile duct carcinomas, an immunohistochemical study for KIT was performed in 289 cases of extrahepatic bile duct carcinomas, and mutational analysis of exon 11 of the c-kit gene was performed in 20 cases that were arbitrarily retrieved from the cases with KIT expression. Cytoplasmic KIT expression was observed in 54 cases (19%) and nuclear KIT in 58 cases (20%) of extrahepatic bile duct carcinoma. Nuclear KIT expression was more frequent in cases with vascular invasion (P<0.001), whereas cytoplasmic KIT expression was more common in tumors of T1-T3 than in those of T4 (P=0.04), and was more frequently observed in cases with a papillary growth pattern (P=0.03). Patients with cytoplasmic KIT-positive tumors had significantly better survival both by univariate (P=0.01) and multivariate analyses (P=0.04). Infrequent cytoplasmic KIT expression without mutation of exon 11 suggests that imatinib mesylate may not be effective for the treatment of extrahepatic bile duct carcinoma. However, immunohistochemical study for KIT may be helpful in routine pathologic examinations for evaluating better prognosis for patients with extrahepatic bile duct carcinoma. In addition, more frequent nuclear expression of KIT in cases with vascular invasion suggests that nuclear KIT expression may contribute to the progression of extrahepatic bile duct carcinoma.
Collapse
Affiliation(s)
- Seung-Mo Hong
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
156
|
Sihto H, Tynninen O, Bützow R, Saarialho-Kere U, Joensuu H. Endothelial cell KIT expression in human tumours. J Pathol 2007; 211:481-8. [PMID: 17294421 DOI: 10.1002/path.2125] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Receptor tyrosine kinases expressed in endothelial cells are potential targets for therapy with specific tyrosine kinase inhibitors. Endothelial cell KIT expression has not been systematically evaluated in human cancer. In the present study, endothelial cell KIT expression was assessed in 345 tumours consisting of 34 different histological types using a tissue microarray technique. Marked KIT expression occurred in the tumour endothelial cells only in primary glioblastomas in the microarray. Moderate to strong KIT and phosphorylated KIT expression was detected in the tumour endothelial cells in six (16%) and seven (19%) of the 37 primary glioblastomas examined, respectively. In whole tissue sections, KIT and phosphorylated KIT were expressed in tumour endothelial cells in 13 (59%) and 11 (50%) of the 22 glioblastomas examined, respectively. RNA in situ hybridization showed KIT mRNA expression in most glioblastomas both in tumour vessel endothelial cells and in perinecrotic palisading glioblastoma cells, whereas little KIT mRNA was found in the endothelial cells of colon or pancreatic carcinomas. Phosphorylated KIT, its ligand stem cell factor, and the downstream signalling molecules phosphorylated Akt and mTOR were often expressed in glioblastoma cells located in the perinecrotic tumour areas that often also contained abundant HIF-1alpha. It is concluded that marked KIT and phosphorylated KIT expression is frequently present in the endothelial cells of glioblastomas, which are known to harbour florid microvascular proliferation with characteristic morphological features. Glioblastomas also express phosphorylated KIT and its activated downstream signalling molecules in the tumour cells. Lower levels of KIT and phosphorylated KIT are present in endothelial cells of other tumour types and in normal tissues. Endothelial cell and tumour cell expression of activated KIT might explain in part the responsiveness of glioblastomas to the combination of imatinib (an inhibitor of KIT) and hydroxyurea.
Collapse
Affiliation(s)
- H Sihto
- Laboratory of Molecular Oncology, Biomedicum Helsinki, Department of Pathology, Helsinki University Central Hospital, Finland.
| | | | | | | | | |
Collapse
|
157
|
Abstract
Sunitinib (SU011248) is an oral small molecular tyrosine kinase inhibitor that exhibits potent antiangiogenic and antitumor activity. Tyrosine kinase inhibitors such as SU6668 and SU5416 (semaxanib) demonstrated poor pharmacologic properties and limited efficacy; therefore, sunitinib was rationally designed and chosen for its high bioavailability and its nanomolar-range potency against the antiangiogenic receptor tyrosine kinases (RTKs)--vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor (PDGFR). Sunitinib inhibits other tyrosine kinases including, KIT, FLT3, colony-stimulating factor 1 (CSF-1), and RET, which are involved in a number of malignancies including small-cell lung cancer, GI stromal tumors (GISTs), breast cancer, acute myelogenous leukemia, multiple endocrine neoplasia types 2A and 2B, and familial medullary thyroid carcinoma. Sunitinib demonstrated robust antitumor activity in preclinical studies resulting not only in tumor growth inhibition, but tumor regression in models of colon cancer, non-small-cell lung cancer, melanoma, renal carcinoma, and squamous cell carcinoma, which were associated with inhibition of VEGFR and PDGFR phosphorylation. Clinical activity was demonstrated in neuroendocrine, colon, and breast cancers in phase II studies, whereas definitive efficacy has been demonstrated in advanced renal cell carcinoma and in imatinib-refractory GISTs, leading to US Food and Drug Administration approval of sunitinib for treatment of these two diseases. Studies investigating sunitinib alone in various tumor types and in combination with chemotherapy are ongoing. The clinical benchmarking of this small-molecule inhibitor of members of the split-kinase domain family of RTKs will lead to additional insights regarding the biology, potential biomarkers, and clinical utility of agents that target multiple signaling pathways in tumor, stromal, and endothelial compartments.
Collapse
Affiliation(s)
- Laura Q M Chow
- Department of Medical Oncology, University of Colorado Health Sciences Center, Aurora, CO 80045, USA
| | | |
Collapse
|
158
|
Abstract
Small-cell lung cancer is a chemo-sensitive disease with a response rate ranging from 70 to 90% for first-line treatment; however, relapses are very common and as a result long-term survival is poor. Chemotherapy has demonstrated a benefit over the best supportive care, even in patients who have relapsed after initial treatment with a platinum-based regimen. Agents currently being used in salvage therapy include topotecan, cyclophosphamide, doxorubicin and vincristine regimen. In the last 5 years, several drugs have shown promise in initial evaluation; however, randomized phase III trials would be needed to answer this question. Our understanding of the biology of small-cell lung cancer has improved dramatically over the past few years and this has translated into the developments of new therapeutic targets for this disease. Agents affecting several targets, including bcl-2, matrix metalloproteinases, epidermal growth factors and angiogenesis, are being studied currently and have the potential to change the treatment paradigms of this otherwise fatal malignancy. This review focuses on the various current and future options, including cytoxic and targeted agents, for salvage therapy in patients with this disease.
Collapse
Affiliation(s)
- Hatem A Azim
- Department of Medical Oncology, National Cancer Institute, Cairo University, Cairo, Egypt
| | | |
Collapse
|
159
|
Abstract
Imatinib (Glivec) is a specific inhibitor of tyrosine kinase receptor, in particular of the proto-oncogene c-kit. Proto-oncogene c-kit is expressed or mutated in stromal digestive tumors (GIST). Pharmacokinetic (PK) analysis showed that imatinib displayed linear PK in patients with advanced GIST. Imatinib is extensively metabolized by the cytochrome P450 enzyme system. Alpha-1-acid glycoprotein (AAG), a protein involved in the acute phase of inflammation, is implicated in protein binding of imatinib and seems to play a key role in imatinib PK.
Collapse
Affiliation(s)
- Catherine Delbaldo
- Service d'oncologie, Hôpital Henri-Mondor, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil Cedex, France.
| |
Collapse
|
160
|
Raut CP, Morgan JA, Ashley SW. Current issues in gastrointestinal stromal tumors: incidence, molecular biology, and contemporary treatment of localized and advanced disease. Curr Opin Gastroenterol 2007; 23:149-58. [PMID: 17268243 DOI: 10.1097/mog.0b013e32802086d0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Few areas in oncology have witnessed the major paradigm shift that has been noted in the understanding and management of gastrointestinal stromal tumors. This review highlights the progress made over the last 2 years. RECENT FINDINGS Population-based studies have provided insight into the true incidence of gastrointestinal stromal tumors. Improved understanding of the molecular biology has provided prognostic implications and may guide treatment in the future. More mature follow-up data from phase III trials have proven that the targeted tyrosine kinase inhibitor imatinib mesylate is a dramatically effective agent, but the duration of its benefits are finite, and drug resistance is an increasingly more common phenomenon. Adjuvant and neoadjuvant trials of imatinib are currently underway. A second targeted tyrosine kinase inhibitor, sunitinib malate, has been approved for the treatment of imatinib-resistant gastrointestinal stromal tumors after recent encouraging results. Finally, the success with imatinib and sunitinib has encouraged investigators to reevaluate the role of surgery in advanced gastrointestinal stromal tumors. SUMMARY The multidisciplinary management of gastrointestinal stromal tumors serves as a model of how new targeted molecular therapies can be combined with traditional treatment modalities to improve survival in advanced malignancies.
Collapse
Affiliation(s)
- Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
161
|
|
162
|
Nigri GR, Dente M, Valabrega S, Aurello P, D'Angelo F, Montrone G, Ercolani G, Ramacciato G. Gastrointestinal stromal tumor of the anal canal: an unusual presentation. World J Surg Oncol 2007; 5:20. [PMID: 17306018 PMCID: PMC1821025 DOI: 10.1186/1477-7819-5-20] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2006] [Accepted: 02/16/2007] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Gastrointestinal stromal tumors (GIST) of the stomach are the most frequent followed by those of the intestinal tract, while colon and rectum represent rare sites. GIST of the anal canal are extremely rare. They have been studied along with GIST of the rectum, as a single entity, and along with them they represent 5% of GIST. GIST arising from the anal canal account for only 2%-8% of the anorectal GIST. Thus anal GIST must be considered an exceptional case. CASE PRESENTATION A 78-year-old man was referred to our Institution for an anal mass, in absence of any symptom. The patient was treated by local excision. An histological diagnosis of a low grade GIST was made. No further treatment was necessary. No local recurrence of distant metastases were found at follow-up. CONCLUSION At the moment, only ten cases of c-kit positive anal GIST are reported in the literature. These few data are not sufficient to establish a widely accepted approach for this neoplasia. We recommend to perform an initial local excision, to define the risk of aggressive behavior and the resection margins and proceed to a more aggressive treatment, if the GIST belongs to high or very high risk group. The role of adjuvant therapy is still uncertain. Although inhibitors of tyrosine-kinase receptor needs further studies before their routine use, their role in case of distant or local recurrence has been accepted. Patients' close follow up is mandatory to disclose as soon as possible local recurrences or metastases.
Collapse
Affiliation(s)
- Giuseppe R Nigri
- Department of Surgery, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Mario Dente
- Department of Surgery, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Stefano Valabrega
- Department of Surgery, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Paolo Aurello
- Department of Surgery, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Francesco D'Angelo
- Department of Surgery, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Giuseppe Montrone
- Department of Pathology, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Giorgio Ercolani
- Department of Surgery, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| | - Giovanni Ramacciato
- Department of Surgery, University of Rome "La Sapienza", 2School of Medicine, Sant'Andrea Hospital, Rome, Italy
| |
Collapse
|
163
|
Stewart DR, Corless CL, Rubin BP, Heinrich MC, Messiaen LM, Kessler LJ, Zhang PJ, Brooks DG. Mitotic recombination as evidence of alternative pathogenesis of gastrointestinal stromal tumours in neurofibromatosis type 1. J Med Genet 2007; 44:e61. [PMID: 17209131 PMCID: PMC2597901 DOI: 10.1136/jmg.2006.043075] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is a neurocutaneous disorder resulting in the growth of a variety of tumours, and is inherited in an autosomal dominant pattern. Gastrointestinal stromal tumours (GISTs) are mesenchymal tumours that commonly harbour oncogenic mutations in KIT or PDGFRA and are thought to arise from the interstitial cells of Cajal (ICC; the pacemaker cells of the gut). AIM To characterise two patients with NF1 and GISTs. METHODS Two patients were genotyped for germline mutations in NF1. GISTs from both patients were genotyped for somatic mutations in KIT and PDGFRA. Loss of heterozygosity (LOH) of NF1 in one GIST was assessed by genotyping seven microsatellite markers spanning 2.39 Mb of the NF1 locus in the tumour and in genomic DNA. The known germline mutation in NF1 was confirmed in GIST DNA by sequencing. The copy number of the mutated NF1 allele was determined by multiplex ligand-dependent probe amplification. RESULTS GISTs from both patients were of wild type for mutations in KIT and PDGFRA. In the GIST with adequate DNA, all seven markers were informative and showed LOH at the NF1 locus; sequencing of NF1 from that GIST showed no wild-type sequence, suggesting that it was lost in the tumour. Multiplex ligand-dependent probe amplification analysis showed that two copies of all NF1 exons were present. CONCLUSIONS This is the first evidence of mitotic recombination resulting in a reduction to homozygosity of a germline NF1 mutation in an NF1-associated GIST. We hypothesise that the LOH of NF1 and lack of KIT and PDGFRA mutations are evidence of an alternative pathogenesis in NF1-associated GISTs.
Collapse
|
164
|
Day AS, Lou PJ, Lin WC, Chou CC. Over-expression of c-kit in a primary leiomyosarcoma of the thyroid gland. Eur Arch Otorhinolaryngol 2007; 264:705-8. [PMID: 17256123 DOI: 10.1007/s00405-007-0242-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Accepted: 12/09/2006] [Indexed: 11/30/2022]
Abstract
Primary leiomyosarcoma of the thyroid gland is rare. In this paper, we report a case of high-grade leiomyosarcoma of the thyroid gland in a 43-year-old man. Lung metastasis was also noted in this patient. Despite of aggressive surgical treatment, the patient died of uncontrolled local recurrent disease 6 months after the initial operation. Immunohistochemical studies showed the tumor cells were positive for c-kit proto-oncogene product. Imatinib mesylate was used as a post-operative adjuvant treatment but the response was poor. The role of tyrosine kinase inhibitors on the treatment of thyroid leiomyosarcomas is still unclear because this is the first report of c-kit over-expression in such tumors. Nevertheless, our results show that c-kit over-expression might not be an indicator of good response to imatinib mesylate treatment in thyroid leiomyosarcomas.
Collapse
Affiliation(s)
- An-Shiou Day
- Department of Otolaryngology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | | | | | | |
Collapse
|
165
|
Corless CL, Harrell P, Lacouture M, Bainbridge T, Le C, Gatter K, White C, Granter S, Heinrich MC. Allele-specific polymerase chain reaction for the imatinib-resistant KIT D816V and D816F mutations in mastocytosis and acute myelogenous leukemia. J Mol Diagn 2007; 8:604-12. [PMID: 17065430 PMCID: PMC1876167 DOI: 10.2353/jmoldx.2006.060089] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Oncogenic mutations of the receptor tyrosine kinase KIT contribute to the pathogenesis of gastrointestinal stromal tumors, systemic mastocytosis (SM), and some cases of acute myelogenous leukemia (AML). The D816V substitution in the activation loop of KIT results in relative resistance to the kinase inhibitor imatinib (Gleevec). Because this mutation occurs in 80 to 95% of adult SM, its detection has diagnostic and predictive significance. Unfortunately, the fraction of mutation-positive cells in clinical SM samples is often below the 20 to 30% threshold needed for detection by direct DNA sequencing. We have developed an allele-specific polymerase chain reaction assay using a mutation-specific primer combined with a wild-type blocking oligonucleotide that amplifies D816V at the level of 1% mutant allele in DNA extracted from formalin-fixed, paraffin-embedded tissue. There were no amplifications among 64 KIT wild-type tumors and cell lines, whereas all D816V-mutant samples (eight AML and 11 mast cell disease) were positive. Other D816 substitutions associated with resistance to imatinib in vitro are rare in SM. Among these D816F was detectable with the assay whereas D816H, D816Y, and D816G did not amplify. Nine biopsies (bone marrow, skin, or colon) with suspected SM were negative by denaturing high performance liquid chromatography and/or DNA sequencing but positive by allele-specific polymerase chain reaction. Thus, the assay may be useful in confirming the diagnosis of SM.
Collapse
Affiliation(s)
- Christopher L Corless
- OHSU Dept. of Pathology (L471), 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Thomas DA. Philadelphia chromosome positive acute lymphocytic leukemia: a new era of challenges. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2007; 2007:435-443. [PMID: 18024662 DOI: 10.1182/asheducation-2007.1.435] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Significant advances in the treatment of Philadelephia chromosome (Ph)- or BCR-ABL-positive acute lymphocytic leukemia (ALL) have been made since the discovery of the selective ABL tyrosine kinase inhibitors (TKIs). Whereas the outcome with standard chemotherapy was previously dismal, incorporation of imatinib mesylate into frontline therapy has improved relapse-free and overall survival. The benefit of imatinib extends particularly to instances where allogeneic stem cell transplantation in first complete remission is prohibited by older age, comorbidities, or lack of a suitable donor. However, the emergence of resistance to imatinib presents new therapeutic challenges. The development of novel TKIs with enhanced inhibitory potency against ABL and other kinases may further improve on the results observed with imatinib. Optimal use of these novel agents in the treatment schema of Ph(+) ALL will be paramount in ensuring continued success in the eradication of this disease. Herein, the new approaches to the management of Ph(+) ALL are reviewed.
Collapse
Affiliation(s)
- Deborah A Thomas
- Anderson Cancer Ctr., 1515 Holcombe Blvd., Unit 428, Houston, TX 77030-1402, USA.
| |
Collapse
|
167
|
Abstract
We report two patients with metastatic gastrointestinal stromal tumor (GIST) who had large subcapsular hepatic hematomas that developed while on imatinib mesylate (Gleevec) therapy. We describe the pertinent radiologic features of the subcapsular hematomas in these patients, and discuss possible etiologies for the bleeding in each patient.
Collapse
|
168
|
Chirieac LR, Trent JC, Steinert DM, Choi H, Yang Y, Zhang J, Patel SR, Benjamin RS, Raymond AK. Correlation of immunophenotype with progression-free survival in patients with gastrointestinal stromal tumors treated with imatinib mesylate. Cancer 2006; 107:2237-44. [PMID: 16998931 DOI: 10.1002/cncr.22226] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND The therapy for gastrointestinal stromal tumor (GIST) has been revolutionized by imatinib mesylate (IM). It is unknown whether the levels of KIT expression or the presence of CD34, smooth muscle actin (SMA), desmin, or S-100 protein predicts patient outcome from IM therapy. In the current study, the prognostic effects for KIT and other proteins (CD34, SMA, desmin, S-100) were analyzed in a series of GISTs in which protein expression was evaluated by immunohistochemical analysis (IHC). METHODS The cases of 106 patients with GIST who were uniformly treated with IM at the study institution between December 15, 2000, and January 13, 2002 were evaluated retrospectively. The association between KIT intensity, CD34, desmin, SMA, S-100 protein, and progression-free survival (PFS) was studied. Kaplan-Meier analysis and the Cox proportional hazards regression model were used for statistical analysis. RESULTS The majority of tumors arose from the stomach (37%), small intestine (35%), and colorectum (14%). KIT expression as determined by IHC was categorized as weak (10%), intermediate (32%), or strong (58%). Patient tumors expressed CD34 (75%), SMA (56%), desmin (1%), and S-100 protein (32%). Patients whose GIST had weak, intermediate, or strong KIT expression were found to have an 18-month PFS rate of 80%, 84%, and 69%, respectively (P = .30). The presence or absence of CD34, SMA, desmin, or S-100 protein did not appear to correlate with PFS after IM. CONCLUSIONS Patients with the appropriate clinical presentation and KIT-positive GIST tumors appear to benefit from IM independent of the level of KIT or the expression of CD34, SMA, desmin, or S-100 protein by IHC.
Collapse
Affiliation(s)
- Lucian R Chirieac
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Katiyar S, Jiao X, Wagner E, Lisanti MP, Pestell RG. Somatic excision demonstrates that c-Jun induces cellular migration and invasion through induction of stem cell factor. Mol Cell Biol 2006; 27:1356-69. [PMID: 17145782 PMCID: PMC1800718 DOI: 10.1128/mcb.01061-06] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cancer cells arise through sequential acquisition of mutations in tumor suppressors and oncogenes. c-Jun, a critical component of the AP-1 complex, is frequently overexpressed in diverse tumor types and has been implicated in promoting cellular proliferation, migration, and angiogenesis. Functional analysis of candidate genetic targets using germ line deletion in murine models can be compromised through compensatory mechanisms. As germ line deletion of c-jun induces embryonic lethality, somatic deletion of the c-jun gene was conducted using floxed c-jun (c-jun(f/f)) conditional knockout mice. c-jun-deleted cells showed increased cellular adhesion, stress fiber formation, and reduced cellular migration. The reduced migratory velocity and migratory directionality was rescued by either c-Jun reintroduction or addition of secreted factors from wild-type cells. An unbiased analysis of cytokines and growth factors, differentially expressed and showing loss of secretion upon c-jun deletion, identified stem cell factor (SCF) as a c-Jun target gene. Immunoneutralizing antibody to SCF reduced migration of wild-type cells. SCF addition rescued the defect in cellular adhesion, cellular velocity, directional migration, transwell migration, and cellular invasion of c-jun(-/-) cells. c-Jun induced SCF protein, mRNA, and promoter activity. Induction of the SCF promoter required the c-Jun DNA-binding domain. c-Jun bound to the SCF promoter in chromatin immunoprecipitation assays. Mutation of the c-Jun binding site abolished c-Jun-mediated induction of the SCF promoter. These studies demonstrate an essential role of c-Jun in cellular migration through induction of SCF.
Collapse
Affiliation(s)
- Sanjay Katiyar
- Departments of Cancer Biology and Medical Oncology, The Kimmel Cancer Center, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA 19107, USA
| | | | | | | | | |
Collapse
|
170
|
Abstract
Response to perioperative chemotherapy or chemoradiation can hardly be predicted on the basis of molecular marker analyses. In contrast, by means of metabolic and molecular imaging using positron emission tomography, response can be assessed as early as 14 days after the start chemotherapy for many tumors. There is no doubt that the prognosis of patients with surgically resected tumors is much better in the case of a response to chemotherapy or chemoradiation. Important consequences can be deduced from this regarding the indications for perioperative therapies, the radicality of surgery or the surgical indications per se. In the following, we delineate the current knowledge on response prediction and early response evaluation in selected tumor entities and draw possible conclusions for clinical practice and future clinical studies.
Collapse
Affiliation(s)
- J R Siewert
- Chirurgische Klinik, Klinikum rechts der Isar, Technische Universität, Ismaninger Strasse 22, 81675, München, Deutschland.
| | | |
Collapse
|
171
|
Izzedine H, Buhaescu I, Rixe O, Deray G. Sunitinib malate. Cancer Chemother Pharmacol 2006; 60:357-64. [PMID: 17136543 DOI: 10.1007/s00280-006-0376-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Accepted: 10/24/2006] [Indexed: 11/25/2022]
Abstract
Recently, there has been a growing interest in understanding the role of receptor tyrosine kinases (RTK), such as vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), stem cell factor receptor (KIT), and fms-like tyrosine kinase 3 (FLT3), in promoting tumor angiogenesis, tumor growth and metastasis. Sunitinib (sunitinib malate; SU11248; SUTENT; Pfizer Inc, New York, NY, USA) is a novel, orally bio-available, oxindole, multi-targeted tyrosine kinase inhibitor with high binding affinity for VEGFR and PDGFR which has shown anti-tumor and anti-angiogenic activities. This drug recently received approval from the US Food and Administration (FDA) in two indications simultaneously: advanced renal cell carcinoma (adRCC) and gastrointestinal stromal tumors (GIST), in patients who are resistant or intolerant to the treatment with imatinib. The present article reviews the recent pharmacologic and clinical data related to the use of this new promising drug in the field of oncology.
Collapse
Affiliation(s)
- Hassane Izzedine
- Department of Nephrology, Pitie-Salpetriere Hospital 83, Blvd de l'Hôpital, 75013 Paris, France.
| | | | | | | |
Collapse
|
172
|
Isotani M, Tamura K, Yagihara H, Hikosaka M, Ono K, Washizu T, Bonkobara M. Identification of a c-kit exon 8 internal tandem duplication in a feline mast cell tumor case and its favorable response to the tyrosine kinase inhibitor imatinib mesylate. Vet Immunol Immunopathol 2006; 114:168-72. [PMID: 16908071 DOI: 10.1016/j.vetimm.2006.07.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Revised: 06/22/2006] [Accepted: 07/07/2006] [Indexed: 11/22/2022]
Abstract
The gain-of-function mutations within c-kit, a protooncogene encoding KIT, induce constitutive ligand-independent kinase activation and are important for the pathogenesis of mast cell proliferative disease in humans as well as in dogs. Despite the clinical importance of feline mast cell tumors, no mutation has been shown within the c-kit gene in cats. In the present report, we analyzed the c-kit nucleotide sequence in the case of a cat that showed systemic mastocytosis and mastocytemia. Within the c-kit cDNA prepared from the malignant mast cells, we identified an 12-bp internal tandem duplication at the region corresponding to exon 8, resulting in a four amino acid insertion between residues Thr418 and His419 within the fifth immunoglobulin-like domain of KIT. The cat underwent therapy with the kinase inhibitor imatinib mesylate (Gleevec) at a dose of 10mg/kg. The tumor masses greatly responded and were undetectable after 5 weeks of treatment. Correspondingly, the number of mast cells in the peripheral blood was markedly reduced. It is, therefore, considered that the internal tandem duplication within the domain contributes to the neoplastic transformation of mast cells in the cat by increasing KIT phosphorylation.
Collapse
Affiliation(s)
- Mayu Isotani
- Department of Veterinary Clinical Pathology, Nippon Veterinary and Life Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602, Japan
| | | | | | | | | | | | | |
Collapse
|
173
|
Hu Y, Swerdlow S, Duffy TM, Weinmann R, Lee FY, Li S. Targeting multiple kinase pathways in leukemic progenitors and stem cells is essential for improved treatment of Ph+ leukemia in mice. Proc Natl Acad Sci U S A 2006; 103:16870-5. [PMID: 17077147 PMCID: PMC1629087 DOI: 10.1073/pnas.0606509103] [Citation(s) in RCA: 229] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
It is generally believed that shutting down the kinase activity of BCR-ABL by imatinib will completely inhibit its functions, leading to inactivation of its downstream signaling pathways and cure of the disease. Imatinib is highly effective at treating human Philadelphia chromosome-positive (Ph(+)) chronic myeloid leukemia (CML) in chronic phase but not Ph(+) B cell acute lymphoblastic leukemia (B-ALL) and CML blast crisis. We find that SRC kinases activated by BCR-ABL remain fully active in imatinib-treated mouse leukemic cells, suggesting that imatinib does not inactivate all BCR-ABL-activated signaling pathways. This SRC pathway is essential for leukemic cells to survive imatinib treatment and for CML transition to lymphoid blast crisis. Inhibition of both SRC and BCR-ABL kinase activities by dasatinib affords complete B-ALL remission. However, curing B-ALL and CML mice requires killing leukemic stem cells insensitive to both imatinib and dasatinib. Besides BCR-ABL and SRC kinases, stem cell pathways must be targeted for curative therapy of Ph(+) leukemia.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- B-Lymphocytes/enzymology
- Benzamides
- Blast Crisis/enzymology
- Burkitt Lymphoma/drug therapy
- Burkitt Lymphoma/enzymology
- Cell Line, Tumor
- Cell Transformation, Neoplastic/metabolism
- Dasatinib
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/enzymology
- Piperazines/therapeutic use
- Protein Kinase Inhibitors/therapeutic use
- Pyrimidines/therapeutic use
- Thiazoles/therapeutic use
- src-Family Kinases/deficiency
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Yiguo Hu
- *The Jackson Laboratory, Bar Harbor, ME 04609; and
| | | | | | | | | | - Shaoguang Li
- *The Jackson Laboratory, Bar Harbor, ME 04609; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
174
|
Appel S, Balabanov S, Brümmendorf TH, Brossart P. Effects of imatinib on normal hematopoiesis and immune activation. Stem Cells 2006; 23:1082-8. [PMID: 16140870 DOI: 10.1634/stemcells.2005-0069] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The selective tyrosine kinase inhibitor imatinib (Glivec; Novartis International, Basel, Switzerland, http://www.glivec.com/content/home.jsp) is increasingly used for the treatment of Philadelphia chromosome-positive leukemias and other malignancies. In principle, the drug is well tolerated and clinical side effects are mostly moderate. However, it was shown that imatinib can affect the function of normal, nonmalignant cells, resulting in myelosuppression in treated patients. Recently, it has been demonstrated that imatinib might affect mobilization, proliferation, and differentiation of hematopoietic progenitor cells while leaving hematopoietic stem cells unaffected. Furthermore, in several in vitro studies and animal models, it was demonstrated that imatinib can affect the function and differentiation of antigen-presenting cells and inhibit the effector functions of T lymphocytes. Moreover, the induction of specific cytotoxic T cells seems to be impaired in chronic myeloid leukemia (CML) patients treated with imatinib compared with patients receiving interferon-alpha. This is of importance because some of the therapeutic effects in the treatment of patients with CML are mediated by the induction of leukemia-specific T-cell responses. Further studies investigating the effects of imatinib on normal hematopoiesis are of interest as they might lead to a better understanding of the clinically observed side effects and also might help identify new therapeutic applications of the drug, possibly in Bcr-Abl-negative myeloproliferative disorders and potentially as an immunomodulatory agent.
Collapse
Affiliation(s)
- Silke Appel
- Department of Hematology, Oncology and Immunology, University of Tübingen, Otfried-Müller Str. 10, D-72076 Tübingen, Germany
| | | | | | | |
Collapse
|
175
|
Litz J, Krystal GW. Imatinib inhibits c-Kit-induced hypoxia-inducible factor-1alpha activity and vascular endothelial growth factor expression in small cell lung cancer cells. Mol Cancer Ther 2006; 5:1415-22. [PMID: 16818499 DOI: 10.1158/1535-7163.mct-05-0503] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular endothelial growth factor (VEGF) is one of the most important mediators of tumor angiogenesis. In addition to hypoxia, peptide growth factors are known to regulate VEGF expression but the effect of stem cell factor (SCF), the ligand for c-Kit, on VEGF expression has not been characterized. We therefore studied the effect of SCF-mediated c-Kit activation on VEGF expression by the H526 small cell lung cancer (SCLC) cell line. SCF treatment doubled VEGF mRNA expression and VEGF secretion in the absence of other exogenous growth factors, an effect efficiently blocked by imatinib. The increase in VEGF mRNA occurred within the first 2 hours of treatment and was not caused by alterations in mRNA stability. The phosphatidylinositol 3-kinase inhibitor LY294002 blocked the increase in VEGF mRNA, implicating c-Kit-mediated activation of phosphatidylinositol 3-kinase in the phenomenon. VEGF promoter-reporter transfections indicated that a SCF-mediated increase in VEGF promoter activity paralleled the increase in VEGF mRNA, documenting that SCF mediated its effects through enhanced VEGF transcription. Mutation of the core hypoxia-inducible factor (HIF)-1 binding element in the VEGF promoter significantly blunted SCF-responsiveness. SCF increased nuclear levels of the HIF-1alpha transcription factor, which correlated well with increased HIF-1alpha binding to a consensus hypoxia-responsive element. SCF-mediated effects on HIF-1alpha expression were additive with those produced by CoCl2, a hypoxia-mimetic agent. These data indicate that activation of c-Kit by SCF leads to a predominantly HIF-1alpha-mediated enhancement of VEGF expression and that inhibition of c-Kit signaling with imatinib could result in inhibition of tumor angiogenesis.
Collapse
Affiliation(s)
- Julie Litz
- Department of Medicine, Virginia Commonwealth University and McGuire Veterans Affairs Medical Center, Richmond, VA, USA
| | | |
Collapse
|
176
|
Yamanaka T, Okamoto T, Ichinose Y, Oda S, Maehara Y. Methodological aspects of current problems in target-based anticancer drug development. Int J Clin Oncol 2006; 11:167-75. [PMID: 16850122 DOI: 10.1007/s10147-006-0580-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Indexed: 01/13/2023]
Abstract
Differently from the conventional antineoplastic agents, target-based drugs are designed a priori, based on our knowledge of various physiological molecules that has been obtained by the development of molecular biology. This "Copernican revolution" in drug development may imply a paradigm shift in this field. However, contrary to the initial expectations, many drugs developed by this approach are now faced with difficulties, mainly because of the fundamental and theoretical limits of this approach. All of the physiological functions are not always known in all target molecules. In low-molecular-weight drugs, i.e., "inhibitors," targets disperse, due to the structural similarities in physiological molecules. This double-faced "out-of-focusing" causes many problems in various steps of drug development, drug design, clinical trials, and administration to patients. Many drugs are now being abandoned because of unexpectedly lower response rates or unforeseeable adverse effects, and the variety of the drugs exhibits a kaleidoscopic appearance. The double-faced "out-of-focusing" derives from the methodological limits in molecular biology, i.e., elementalism, and limits in our techniques for drug development. To overcome these currently inevitable limits, it appears essential to elucidate the specific changes in target molecules that chiefly promote tumor growth and, consequently, strongly predict response to the administered drugs. Precise and efficient detection of responder populations is the key to the development and establishment of target-based anticancer therapies.
Collapse
Affiliation(s)
- Takeharu Yamanaka
- Cancer Statistics Laboratory, Institute for Clinical Research, National Kyushu Cancer Center, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
177
|
Babina M, Rex C, Guhl S, Thienemann F, Artuc M, Henz BM, Zuberbier T. Baseline and stimulated turnover of cell surface c-Kit expression in different types of human mast cells. Exp Dermatol 2006; 15:530-7. [PMID: 16761962 DOI: 10.1111/j.1600-0625.2006.00446.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The receptor tyrosine kinase c-Kit is fundamental to mast cell (MC) development and maintenance. Its regulation can occur at various levels, but nothing is known about how this is accomplished in normal human tissue MC. Likewise, the baseline turnover of c-Kit has not been addressed yet. We used mature MC from human skin, along with the MC lines LAD-2 and HMC-1 and treated them with stem cell factor (SCF), cycloheximide, actinomycin D (AD) and combinations thereof, and determined expression levels of c-Kit and other surface receptors by flow cytometry. Ligand-induced internalization of c-Kit was found to be a universal mechanism and detectable in all MC subtypes. By Western blot analysis of LAD-2 cells, c-Kit was found to nearly disappear 3 h after the addition of SCF to slowly recover thereafter. Investigations into the baseline turnover of c-Kit expression revealed that c-Kit is strongly affected by the inhibition of de novo translation in all MC subsets, while a suppression of transcription had a weaker effect and displayed greater cell-to-cell variation. Only a minor impact on other cell surface receptors (CD29, CD50 and CD54) was noted. On combined treatment, cycloheximide, AD and SCF displayed additive effects, resulting in a complete disappearance of c-Kit from the cell surface. In conclusion, c-Kit represents a rapidly cycling cell surface receptor. It is not only immediately internalized upon binding of its ligand, but it is also heavily affected by the inhibition of translation or transcription when viewed against an average background. Interestingly, c-Kit regulation seems largely independent of the MC subtype.
Collapse
Affiliation(s)
- Magda Babina
- Department of Dermatology and Allergy, Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
178
|
Willmore-Payne C, Holden JA, Chadwick BE, Layfield LJ. Detection of c-kit exons 11- and 17-activating mutations in testicular seminomas by high-resolution melting amplicon analysis. Mod Pathol 2006; 19:1164-9. [PMID: 16741525 DOI: 10.1038/modpathol.3800623] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A subgroup of testicular seminomas has been reported to contain activating mutations in KIT, the transmembrane tyrosine kinase receptor encoded by the c-kit gene. Most mutations are in exon 17, although exon 11-activating mutations have recently been described. For patients refractory to standard therapeutic protocols for seminoma, the presence of c-kit-activating mutations in some of these neoplasms might suggest an alternative therapy with KIT targeting drugs. We used the novel mutation scanning technique, high-resolution melting amplicon analysis, to screen a series of 22 testicular seminomas for c-kit-activating mutations. Four cases (18%) had exon 17-activating mutations and these included D816Y, D816V, Y823N and one case that contained both D816E and D820H. A single case (5%) had an exon 11-activating mutation. Interestingly, the exon 11-activating mutation was L576P, the same mutation that characterizes the rare c-kit mutation-positive cases of malignant melanoma. Fluorescence in situ hybridization (FISH) for c-kit suggested that most seminomas are probably polysomic for c-kit and there was not a significant difference in c-kit FISH characteristics between the mutation-positive and mutation-negative cases. The use of high-resolution melting amplicon analysis as a screening technique will allow for the rapid identification of patients with testicular seminomas whose tumors contain c-kit-activating mutations. This could benefit patients whose tumors are refractory to standard therapeutic protocols.
Collapse
Affiliation(s)
- Carlynn Willmore-Payne
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, UT 84132, USA
| | | | | | | |
Collapse
|
179
|
Pan J, Quintás-Cardama A, Kantarjian HM, Akin C, Manshouri T, Lamb P, Cortes JE, Tefferi A, Giles FJ, Verstovsek S. EXEL-0862, a novel tyrosine kinase inhibitor, induces apoptosis in vitro and ex vivo in human mast cells expressing the KIT D816V mutation. Blood 2006; 109:315-22. [PMID: 16912224 DOI: 10.1182/blood-2006-04-013805] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Gain-of-function mutations of the receptor tyrosine kinase KIT play a key role in the pathogenesis of systemic mastocytosis (SM), gastrointestinal stromal tumors (GISTs), and some cases of acute myeloid leukemia (AML). Whereas KIT juxtamembrane domain mutations seen in most patients with GIST are highly sensitive to imatinib, the kinase activation loop mutant D816V, frequently encountered in SM, hampers the binding ability of imatinib. We investigated the inhibitory activity of the novel tyrosine kinase inhibitor EXEL-0862 against 2 subclones of human mast cell line-1 (HMC-1)-HMC-1.1, harboring the juxtamembrane domain mutation V560G, and HMC-1.2, carrying V560G and the activation loop mutation D816V, found in more than 80% of patients with SM. EXEL-0862 inhibited the phosphorylation of KIT in a dose-dependent manner and decreased cell proliferation in both mast cell lines with higher activity against HMC-1.2 cells. The phosphorylation of KIT-dependent signal transducer and activator of transcription-3 (STAT3) and STAT5 was abrogated upon exposure to nanomolar concentrations of EXEL-0862. In addition, EXEL-0862 induced a time- and dose-dependent proapoptotic effect in both mast cell lines and caused a significant reduction in mast-cell content in bone marrow samples from patients with SM harboring D816V and from those without the D816V mutation. We conclude that EXEL-0862 is active against KIT activation loop mutants and is a promising candidate for the treatment of patients with SM and other KIT-driven malignancies harboring active site mutations.
Collapse
Affiliation(s)
- Jingxuan Pan
- The University of Texas M D Anderson Cancer Center, Unit 428, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Potapova O, Laird AD, Nannini MA, Barone A, Li G, Moss KG, Cherrington JM, Mendel DB. Contribution of individual targets to the antitumor efficacy of the multitargeted receptor tyrosine kinase inhibitor SU11248. Mol Cancer Ther 2006; 5:1280-9. [PMID: 16731761 DOI: 10.1158/1535-7163.mct-03-0156] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent achievements in the development of multitargeted molecular inhibitors necessitate a better understanding of the contribution of activity against individual targets to their efficacy. SU11248, a small-molecule inhibitor targeting class III/V receptor tyrosine kinases, including the platelet-derived growth factor (PDGF) and vascular endothelial growth factor (VEGF) receptors, KIT and FLT3, exhibits direct effects on cancer cells as well as antiangiogenic activity. Here, we investigated the contributions of inhibiting individual SU11248 target receptors to its overall antitumor efficacy in tumor models representing diverse signaling paradigms. Consistent with previous results, SU11248 was highly efficacious (frequently cytoreductive) in all models tested. To elucidate the specific contributions of inhibition of PDGF and VEGF receptors to the in vivo efficacy of SU11248, we employed two selective inhibitors, SU10944 (VEGF receptor inhibitor) and Gleevec (PDGF receptor inhibitor). SU10944 alone induced a tumor growth delay in all models evaluated, consistent with a primarily antiangiogenic mode of action. In contrast, Gleevec resulted in modest growth inhibition in tumor models in which the cancer cells expressed its targets (PDGFRbeta and KIT), but was not efficacious against tumors not driven by these target receptor tyrosine kinases. Strikingly, in all but one tumor model evaluated, the antitumor efficacy of SU10944 combined with Gleevec was similar to that of single-agent SU11248, and was greatly superior to that of each compound alone, indicating that the antitumor potency of SU11248 in these models stems from combined inhibition of both PDGF and VEGF receptors. The one exception was a model driven by an activated mutant of FLT3, in which the activity of SU11248, which targets FLT3, was greater than that of SU10944 plus Gleevec. Moreover, SU10944 combined with Gleevec inhibited tumor neoangiogenesis to an extent comparable to that of SU11248. Thus, the potent efficacy of SU11248 in models representing diverse signaling paradigms results from simultaneous inhibition of individual target receptors expressed both in cancer cells and in the tumor neovasculature, supporting the hypothesis that multitargeted inhibitors have the cumulative antitumor efficacy of combined single-target inhibitors.
Collapse
Affiliation(s)
- Olga Potapova
- Cureline, Inc., 393 East Grand Avenue, Suite I, South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | | | |
Collapse
|
181
|
Giuliano CJ, Freemantle SJ, Spinella MJ. Testicular Germ Cell Tumors: A Paradigm for the Successful Treatment of Solid Tumor Stem Cells. CURRENT CANCER THERAPY REVIEWS 2006; 2:255-270. [PMID: 24482633 PMCID: PMC3904303 DOI: 10.2174/157339406777934681] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Treatment of testicular germ cell tumors (TGCTs) has been a success primarily due to the exquisite responsiveness of this solid tumor to cisplatin-based therapy. Despite the promise of cure for the majority of TGCT patients, the effectiveness of therapy for some patients is limited by toxicity and the problem of resistance. There is compelling rationale to further understand the biology of TGCTs in order to better treat other solid tumors and to address the shortcomings of present TGCT therapies. TGCTs contain undifferentiated pluripotent stem cells, known as embryonal carcinoma, that share many properties with human embryonic stem cells. The importance of cancer stem cells in the initiation, progression and treatment of solid tumors is beginning to emerge. We discuss TGCTs in the context of solid tumor curability and targeted cancer stem cell therapy.
Collapse
Affiliation(s)
- Caryl J. Giuliano
- Department of Pharmacology and Toxicology, Dartmouth Medical School, and the Norris Cotton Cancer Center, Dartmouth Hitchcock-Medical Center, Hanover, NH 03755, USA
| | - Sarah J. Freemantle
- Department of Pharmacology and Toxicology, Dartmouth Medical School, and the Norris Cotton Cancer Center, Dartmouth Hitchcock-Medical Center, Hanover, NH 03755, USA
| | - Michael J. Spinella
- Department of Pharmacology and Toxicology, Dartmouth Medical School, and the Norris Cotton Cancer Center, Dartmouth Hitchcock-Medical Center, Hanover, NH 03755, USA
| |
Collapse
|
182
|
Ishikubo T, Akagi K, Kurosumi M, Yamaguchi K, Fujimoto T, Sakamoto H, Tanaka Y, Ochiai A. Immunohistochemical and Mutational Analysis of c-kit in Gastrointestinal Neuroendocrine Cell Carcinoma. Jpn J Clin Oncol 2006; 36:494-8. [PMID: 16844734 DOI: 10.1093/jjco/hyl061] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Gastrointestinal neuroendocrine cell carcinoma (NEC) is a highly aggressive tumor with poor prognosis, for which an effective therapy is highly desirable. Recently, use of a c-kit inhibitor achieved excellent results against gastrointestinal stromal tumor (GIST) that showed c-kit overexpression and mutation in most cases. According to recent studies, 17-44% of pulmonary NEC also expressed c-kit and the antitumor effect of c-kit inhibitor was demonstrated in vitro against small cell carcinoma of the lung. As gastrointestinal NECs are clinicopathologically similar to pulmonary NECs, we investigated c-kit expression and mutation in gastrointestinal NEC. METHODS Surgically resected gastrointestinal NEC was examined for c-kit expression by immunohistochemistry and RT-PCR. Mutation of the c-kit gene was also investigated by means of single-strand conformation polymorphisms (SSCP). RESULTS Twenty-six percent (6 out of 23 patients) of gastrointestinal NEC expressed c-kit protein. c-kit protein expression was demonstrated in 1 out of 4 colorectal, 1 out of 2 duodenal, 4 out of 16 gastric and no esophageal (sample size of 1) NECs. The results of immunohistochemistry for c-kit were consistent with the RT-PCR. No c-kit gene mutation was found in gastrointestinal NEC. CONCLUSION The frequency of c-kit expression in gastrointestinal NEC was similar to that previously reported for pulmonary NEC. These findings suggest that c-kit inhibitor may be effective against some gastrointestinal NECs.
Collapse
Affiliation(s)
- Tsutomu Ishikubo
- Division of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Kitaadachigun, Saitama 362-0806, Japan
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Wyman K, Atkins MB, Prieto V, Eton O, McDermott DF, Hubbard F, Byrnes C, Sanders K, Sosman JA. Multicenter Phase II trial of high-dose imatinib mesylate in metastatic melanoma: significant toxicity with no clinical efficacy. Cancer 2006; 106:2005-11. [PMID: 16565971 DOI: 10.1002/cncr.21834] [Citation(s) in RCA: 239] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Systemic treatment of metastatic melanoma is largely ineffective and alternative approaches are needed. Imatinib mesylate is an oral tyrosine kinase inhibitor that targets bcr-Abl, c-kit, platelet-derived growth factor receptor (PDGFR)-alpha, and PDGFR-beta, leading to remarkable clinical responses in several cancers. Signal transduction via c-kit, PDGFR-alpha, and PDGFR-beta has been demonstrated in malignant melanoma. METHODS The primary objective of this Phase II study was to determine the response rate, response duration, and the frequency of 6-month progression-free survival in patients who could receive up to 2 prior therapeutic regimens. Initially, patients received imatinib at at dose of 400 mg twice orally each day. Based on Simon's optimal design, the study allowed entry of 21 patients; if there were > or = 2 objective responses, accrual would then continue to a total of 41 patients. RESULTS Twenty-six patients were enrolled. Patients experienced 29 episodes of Grade 3 and 2 episodes of Grade 4 toxicity (according to National Cancer Institute common toxicity criteria). No objective clinical responses were noted among the 25 evaluable patients. The median time to progression was 54 days and the median overall survival was 200 days. No patient was free of disease progression at 6 months. Paraffin-embedded tumor specimens from 15 patients were tested for expression of imatinib responsive kinases by immunohistochemistry. Three tumors had moderate and 5 tumors had weak staining for c-kit. Five tumor samples had weak staining for PDGFR-alpha and -beta. CONCLUSIONS Imatinib is an inactive single agent in metastatic melanoma in a population of predominantely pretreated patients. The levels of c-kit and/or PDGFR-alpha, -beta expression in the current study were lower than previously reported. Alternative treatment strategies remain a priority for patients with advanced melanoma.
Collapse
Affiliation(s)
- Ken Wyman
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Wanebo HJ, Argiris A, Bergsland E, Agarwala S, Rugo H. Targeting growth factors and angiogenesis; using small molecules in malignancy. Cancer Metastasis Rev 2006; 25:279-92. [PMID: 16770540 DOI: 10.1007/s10555-006-8508-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Targeted biologic therapy for cancer has evolved from the laboratory to active clinical protocols and applied clinical practice in selected patients. Major targets include epidermal growth factor, and vascular endothelial growth factor receptors which are commonly expressed in gastro-intestinal cancers head & neck and lung cancers, and to some degree breast and gynecologic malignancy. Down stream signal transduction pathway inhibition of B-raf and N-ras mutations are examined in melanoma. New approaches involving re-packaging of chemotherapeutic agents are being exemplified in the nanoparticle formulation of paclitaxel which provides increased access to endothelial and tumor cells with potential enhanced therapeutic efficacy compared to the conventional version solubilized in a cremophor.
Collapse
Affiliation(s)
- Harold J Wanebo
- Department of Surgery, Division Surgical Oncology, Roger Williams Medical Center, Providence, RI, USA.
| | | | | | | | | |
Collapse
|
185
|
Carter CA, Chen C, Brink C, Vincent P, Maxuitenko YY, Gilbert KS, Waud WR, Zhang X. Sorafenib is efficacious and tolerated in combination with cytotoxic or cytostatic agents in preclinical models of human non-small cell lung carcinoma. Cancer Chemother Pharmacol 2006; 59:183-95. [PMID: 16724239 DOI: 10.1007/s00280-006-0257-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2006] [Accepted: 04/23/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE Sorafenib tosylate (sorafenib, BAY 43-9006, Nexavar) is a multi-kinase inhibitor that targets tumor cell proliferation and angiogenesis. These studies evaluated the efficacy and tolerability of combinations of sorafenib plus agents used to treat non-small cell lung cancer (NSCLC) using preclinical models of that disease. METHODS Intravenous (iv) vinorelbine and interperitoneal (ip) cisplatin were administered intermittently (q4d x 3) in combination with sorafenib administered orally (po) once daily for 9 days starting on the same day as the standard agent. In studies with sorafenib and gefitinib, both agents were administered po daily for 10 days starting on the same day. Treatment in all studies was initiated against established sc tumors, and each study was conducted in duplicate. Efficacy was assessed as the delay in tumor growth to a specified size (TGD). RESULTS Vinorelbine (6.7 mg/kg) and sorafenib (40 mg/kg) produced TGDs of 2.4 and 7.8 days, respectively, in the NCI-H460 NSCLC model. Combination therapy produced a 10.0-day TGD with no increase in toxicity. Combination therapy in the NCI-H23 NSCLC model with the highest evaluated dose levels of sorafenib plus cisplatin was well tolerated and produced TGDs equivalent to those produced by cisplatin alone. Lower dose levels of each agent produced approximately additive TGD's. Combination therapy in the A549 NSCLC model with sorafenib and gefitinib produced TGDs equivalent to that produced by sorafenib alone with no toxicity. Tumor growth in the MDA-MB-231 mammary tumor model, that contains mutations in signal transduction proteins downstream of the EGF receptor (the target of gefitinib) was also inhibited by sorafenib, but not by gefitinib. CONCLUSION Concurrent administration of sorafenib and vinorelbine, cisplatin or gefitinib was at least as efficacious as the individual agents alone and was well tolerated. These results support the inclusion of sorafenib in clinical trials in NSCLC employing combinations of both cytotoxic and cytostatic agents.
Collapse
|
186
|
Abstract
The emergence of tumour-specific, molecularly targeted agents signifies a paradigm shift in cancer therapy, with less reliance on drugs that non-discriminately kill tumour and host cells. Although the diversity of targets giving rise to this new generation of anticancer drugs has expanded, many challenges persist in the design of effective treatment regimens. The complex interplay of signal-transduction pathways further complicates the customization of cancer treatments to target single mechanisms. However, despite uncertainty over precise or dominant mechanisms of action, especially for compounds targeting multiple gene products, emerging agents are producing significant therapeutic advances against a broad range of human cancers.
Collapse
Affiliation(s)
- Judith S Sebolt-Leopold
- Pfizer Global Research and Development, Michigan Laboratories, 2800 Plymouth Road, Ann Arbor, Michigan 48105, USA
| | | |
Collapse
|
187
|
Towu E, Stanton M. Gastrointestinal stromal tumour presenting with severe bleeding: a review of the molecular biology. Pediatr Surg Int 2006; 22:462-4. [PMID: 16463034 DOI: 10.1007/s00383-006-1636-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Indexed: 01/25/2023]
Abstract
Gastrointestinal stromal tumours (GIST) are rare in children. A 7-year-old boy presented acutely with a severe upper gastrointestinal bleed and was found following angiography to have such a tumour in the duodenum, which was resected at laparotomy. The presence of CD 117 positive immunostaining was a confirmatory diagnostic marker. The prognosis and underlying molecular biology of the tumour is discussed. Understanding of the molecular pathogenesis has given rise to promising new therapies.
Collapse
Affiliation(s)
- E Towu
- Department of Paediatric Surgery, Lewisham University Hospital NHS Trust, Lewisham High Street, London, SE13 6LH, UK
| | | |
Collapse
|
188
|
Hassan I, You YN, Dozois EJ, Shayyan R, Smyrk TC, Okuno SH, Donohue JH. Clinical, pathologic, and immunohistochemical characteristics of gastrointestinal stromal tumors of the colon and rectum: implications for surgical management and adjuvant therapies. Dis Colon Rectum 2006; 49:609-15. [PMID: 16552495 DOI: 10.1007/s10350-006-0503-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION This study was designed to review the clinical characteristics of surgically treated gastrointestinal stromal tumors of the colon and rectum, evaluate their immunohistochemical and pathologic features based on the current National Institutes of Health criteria, and correlate clinicopathologic findings with the subsequent clinical course. METHODS Patient and disease characteristics at presentation, pathologic features, surgical management, and clinical outcomes of 18 patients with gastrointestinal stromal tumors (4 colon and 14 rectum) diagnosed and primarily treated at our institution between 1979 and 2004 were evaluated. RESULTS Tumors were classified on basis of size and mitotic rate according to current National Institutes of Health recommendations: 67 percent (n = 12) were high-risk, 5 percent (n = 1) were intermediate-risk, 17 percent (n = 3) were low-risk, and 11 percent (n = 2) were very low-risk gastrointestinal stromal tumors. Fifteen of 18 tumors were KIT-positive. The three KIT-negative tumors were platelet-derived growth factor receptor alpha positive. All patients with colonic gastrointestinal stromal tumors (n = 4) underwent segmental resection, whereas patients with rectal gastrointestinal stromal tumors had local excision (n = 5) or radical resection (n = 9). Sixty-six percent (8/12) of patients with high-risk colorectal gastrointestinal stromal tumors developed metastases. None of the patients (n = 6) with intermediate-risk, low-risk, or very low-risk gastrointestinal stromal tumors died of their disease after a median follow-up of 65 (range, 15-266) months. CONCLUSIONS The majority of gastrointestinal stromal tumors of the colon and rectum are high-risk. Patients with high-risk colorectal gastrointestinal stromal tumors have a significant likelihood of developing metastases that is associated with poor prognosis. These patients need to be closely followed for an extended period and should be considered for adjuvant therapy with tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Imran Hassan
- Division of Colon and Rectal Surgery, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
189
|
Dang I, Nelson JK, DeVries GH. c-Kit receptor expression in normal human Schwann cells and Schwann cell lines derived from neurofibromatosis type 1 tumors. J Neurosci Res 2006; 82:465-71. [PMID: 16235251 DOI: 10.1002/jnr.20648] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The growth factor receptor c-Kit has several well-characterized functions during the development of numerous cell types, including red blood cells, mast cells, and melanocytes. Its role in Schwann cells has been described in transformed cells derived from malignant peripheral nerve sheath tumors from patients with neurofibromatosis type 1 (NF1 MPNST; Badache et al. [1998] Oncogene 17:795-800). However, c-Kit functions have not been investigated in normal Schwann cells. We report here that neonatal rat Schwann cells express low c-Kit levels, whereas expression levels for c-Kit are high for Schwann cells derived from MPNST of NF1 patients. In addition, c-Kit expression is not detectable in normal adult human Schwann cells. Although the c-Kit ligand stem cell factor (SCF) induces the phosphorylation of protein kinase B (or Akt) and prevents apoptosis in Schwann cells, SCF has no effect on the proliferation or differentiation of Schwann cells.
Collapse
Affiliation(s)
- Ian Dang
- Research Service, Hines VA Hospital, Hines, IL 60141, USA
| | | | | |
Collapse
|
190
|
Rijn MVD, Fletcher JA. GENETICS OF SOFT TISSUE TUMORS. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2006; 1:435-66. [DOI: 10.1146/annurev.pathol.1.110304.100052] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Matt van de Rijn
- Department of Pathology, Stanford University Medical Center, Stanford, California 94305;
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115;
| |
Collapse
|
191
|
Abstract
Coexpression of Kit ligand and c-kit has been reported in some gynecologic tumors. To determine whether imatinib mesylate is useful in ovarian epithelial tumors, we performed immunohistochemical and mutational analysis. The cases consisted of 33 cases, which included 13 serous cystadenocarcinomas, 1 borderline serous tumor, 8 mucinous cystadenocarcinomas, 6 borderline mucinous tumors and 5 clear cell carcinomas. Five cases of serous cystadenoma and 5 cases of mucinous cystadenoma were also included. In the immunohistochemical study, 3 cases (3/6, 50%) of borderline mucinous cystic tumor and two cases (2/8, 25%) of mucinous cystadenocarcinoma show positive staining for KIT protein. Only one case (1/13, 7.7%) of serous cystadenocarcinoma had positive staining. On mutational analysis, no mutation was identified at exon 11. However, two cases of borderline mucinous tumors and one case of mucinous cystadenocarcinoma had mutations at exon 17. In these cases, the immunohistochemistry also shows focal positive staining at epithelial component. Although, KIT protein expression showed higher incidence in mucinous tumors than serous tumors, they lack KIT-activating mutations in exon 11. Thus, ovarian surface epithelial tumors are unlikely to respond to imatinib mesylate.
Collapse
MESH Headings
- Adult
- Aged
- Cystadenocarcinoma, Mucinous/genetics
- Cystadenocarcinoma, Mucinous/metabolism
- Cystadenocarcinoma, Mucinous/pathology
- Cystadenoma, Mucinous/genetics
- Cystadenoma, Mucinous/metabolism
- Cystadenoma, Mucinous/pathology
- Cystadenoma, Serous/genetics
- Cystadenoma, Serous/metabolism
- Cystadenoma, Serous/pathology
- DNA Mutational Analysis
- DNA, Neoplasm/chemistry
- DNA, Neoplasm/genetics
- Epithelial Cells/chemistry
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Middle Aged
- Mutation
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Polymerase Chain Reaction
- Polymorphism, Single-Stranded Conformational
- Proto-Oncogene Proteins c-kit/biosynthesis
- Proto-Oncogene Proteins c-kit/genetics
Collapse
Affiliation(s)
- Dong-Ja Kim
- Department of Pathology, Fatima Hospital, Daegu, Korea.
| | | | | | | |
Collapse
|
192
|
Garton AJ, Crew APA, Franklin M, Cooke AR, Wynne GM, Castaldo L, Kahler J, Winski SL, Franks A, Brown EN, Bittner MA, Keily JF, Briner P, Hidden C, Srebernak MC, Pirrit C, O'Connor M, Chan A, Vulevic B, Henninger D, Hart K, Sennello R, Li AH, Zhang T, Richardson F, Emerson DL, Castelhano AL, Arnold LD, Gibson NW. OSI-930: A Novel Selective Inhibitor of Kit and Kinase Insert Domain Receptor Tyrosine Kinases with Antitumor Activity in Mouse Xenograft Models. Cancer Res 2006; 66:1015-24. [PMID: 16424037 DOI: 10.1158/0008-5472.can-05-2873] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OSI-930 is a novel inhibitor of the receptor tyrosine kinases Kit and kinase insert domain receptor (KDR), which is currently being evaluated in clinical studies. OSI-930 selectively inhibits Kit and KDR with similar potency in intact cells and also inhibits these targets in vivo following oral dosing. We have investigated the relationships between the potency observed in cell-based assays in vitro, the plasma exposure levels achieved following oral dosing, the time course of target inhibition in vivo, and antitumor activity of OSI-930 in tumor xenograft models. In the mutant Kit-expressing HMC-1 xenograft model, prolonged inhibition of Kit was achieved at oral doses between 10 and 50 mg/kg and this dose range was associated with antitumor activity. Similarly, prolonged inhibition of wild-type Kit in the NCI-H526 xenograft model was observed at oral doses of 100 to 200 mg/kg, which was the dose level associated with significant antitumor activity in this model as well as in the majority of other xenograft models tested. The data suggest that antitumor activity of OSI-930 in mouse xenograft models is observed at dose levels that maintain a significant level of inhibition of the molecular targets of OSI-930 for a prolonged period. Furthermore, pharmacokinetic evaluation of the plasma exposure levels of OSI-930 at these effective dose levels provides an estimate of the target plasma concentrations that may be required to achieve prolonged inhibition of Kit and KDR in humans and which would therefore be expected to yield a therapeutic benefit in future clinical evaluations of OSI-930.
Collapse
Affiliation(s)
- Andrew J Garton
- OSI Pharmaceuticals Inc., 1 Bioscience Park Drive, Farmingdale, NY 11735, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Leithner A, Gapp M, Radl R, Pascher A, Krippl P, Leithner K, Windhager R, Beham A. Immunohistochemical analysis of desmoid tumours. J Clin Pathol 2006; 58:1152-6. [PMID: 16254103 PMCID: PMC1770757 DOI: 10.1136/jcp.2005.026278] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND/AIMS Although the standard treatment for desmoid tumours is complete surgical resection with wide margins, the optimal adjuvant treatment for recurrent or inoperable disease is unclear, often being based on sporadic immunohistochemical reports with a low number of cases. Therefore, a large immunohistochemical study was performed, to provide a theoretical basis for adjuvant treatment regimens. METHODS One hundred and sixteen tissue samples from 80 patients (49 female, 31 male; mean age, 34 years; range, 0-83) with desmoid tumours (46 extra-abdominal, 21 abdominal, 13 intra-abdominal) were tested for oestrogen receptors alpha and beta, progesterone and androgen receptors, and somatostatin, in addition to HER2, cathepsin D, Ki-67, and c-KIT by immunohistochemistry. RESULTS All samples were negative for oestrogen receptor alpha, HER2, and the progesterone receptor. Positive staining for the androgen receptor was found in six extra-abdominal cases. Staining for oestrogen receptor beta was positive in four extra-abdominal, two abdominal, and one intra-abdominal case. Staining for somatostatin was positive in six extra-abdominal, two abdominal, and one intra-abdominal case, and staining for cathepsin D was positive in all cases. Positive staining for Ki-67 was found in 14 extra-abdominal, three abdominal, and three intra-abdominal cases. C-KIT was detectable in one abdominal case only. CONCLUSIONS The data from this immunohistochemical study show that the published effects of antioestrogens and imatinib mesylate in the treatment of aggressive fibromatoses may not be attributable to oestrogen receptor alpha or c-KIT expression.
Collapse
MESH Headings
- Abdominal Neoplasms/chemistry
- Abdominal Neoplasms/drug therapy
- Abdominal Neoplasms/pathology
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/analysis
- Cathepsin D/analysis
- Chemotherapy, Adjuvant
- Child
- Child, Preschool
- Female
- Fibromatosis, Aggressive/drug therapy
- Fibromatosis, Aggressive/metabolism
- Fibromatosis, Aggressive/pathology
- Humans
- Infant
- Infant, Newborn
- Ki-67 Antigen/analysis
- Male
- Middle Aged
- Neoplasm Proteins/analysis
- Proto-Oncogene Proteins c-kit/analysis
- Receptors, Androgen/analysis
- Receptors, Estrogen/analysis
- Receptors, Progesterone/analysis
- Soft Tissue Neoplasms/chemistry
- Soft Tissue Neoplasms/drug therapy
- Soft Tissue Neoplasms/pathology
- Somatostatin/analysis
Collapse
Affiliation(s)
- A Leithner
- Department of Orthopaedic Surgery, Medical University Graz, Auenbruggerplatz 5, A-8036 Graz, Austria
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Kim YC, Kim KS. Drugs for Lung Cancer Treatment. Tuberc Respir Dis (Seoul) 2006. [DOI: 10.4046/trd.2006.60.2.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Young-Chul Kim
- Division of Pulmonology, Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Korea
| | - Kyu-Sik Kim
- Division of Pulmonology, Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Korea
| |
Collapse
|
195
|
Li L, Blumenthal DK, Masaki T, Terry CM, Cheung AK. Differential effects of imatinib on PDGF-induced proliferation and PDGF receptor signaling in human arterial and venous smooth muscle cells. J Cell Biochem 2006; 99:1553-63. [PMID: 16817200 DOI: 10.1002/jcb.20993] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Platelet-derived growth factor (PDGF) has been implicated in smooth muscle cell (SMC) proliferation, a key event in the development of myointimal hyperplasia in vascular grafts. Recent evidence suggests that the PDGF receptor (PDGFR) tyrosine kinase inhibitor, imatinib, can prevent arterial proliferative diseases. Because hyperplasia is far more common at the venous anastomosis than the arterial anastomosis in vascular grafts, we investigated whether imatinib also inhibited venous SMC (VSMC) proliferation, and examined possible differences in its mechanism of action between VSMC and arterial SMC (ASMC). Human ASMC and VSMC were stimulated with PDGF-AB, in the presence or absence of imatinib (0.1-10 microM). Proliferation was assayed using the 5-bromo-2'-deoxyuridine (BrdU) incorporation assay, while PDGFR, Akt and ERK1/2-mitogen activated protein kinase (MAPK) signaling pathways were investigated by immunoblotting. The proliferative response to PDGF at 50 and 100 ng/ml was 32 and 43% greater, respectively, in VSMC than in ASMC. Similarly, PDGF-stimulated proliferation was more sensitive to inhibition by imatinib in VSMC than ASMC (IC(50) = 0.05 microM vs. 0.4 microM; P < 0.01). Imatinib also more effectively inhibited PDGF-induced phosphorylation of PDGFRbeta and Akt in VSMC, compared to ASMC. These data highlight inherent pharmacodynamic differences between VSMC and ASMC in receptor and cell signaling functions and suggest that imatinib therapy may be useful for the prevention of venous stenosis in vascular grafts.
Collapse
Affiliation(s)
- Li Li
- Department of Medicine, University of Utah, 85 North Medical Drive East, Salt Lake City, UT 84112, USA
| | | | | | | | | |
Collapse
|
196
|
Auewarakul CU, Lauhakirti D, Promsuwicha O, Munkhetvit C. C-kit receptor tyrosine kinase (CD117) expression and its positive predictive value for the diagnosis of Thai adult acute myeloid leukemia. Ann Hematol 2005; 85:108-12. [PMID: 16320053 DOI: 10.1007/s00277-005-0039-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2005] [Accepted: 07/30/2005] [Indexed: 12/16/2022]
Abstract
We examined the expression of c-kit receptor tyrosine kinase in 195 Thai adult patients with acute leukemia and determined its specificity and predictive values for the diagnosis of adult acute myeloid leukemia (AML). CD117 was used to detect c-kit expression on CD45 and side-scatter-gated blast cells by flow cytometry. Of 163 AML cases, 67% expressed CD117. None of acute lymphoid leukemia (ALL) had CD117 expression, except one case of T-ALL. The majority of AML patients carrying t(8;21), inv(16), and t(15;17) had high CD117 expression. High proportion of AML cases without c-kit expressed monocytic markers. Significant associations between CD117 and CD34 (P<0.001), CD13 (P=0.006), CD7 (P=0.034), and CD19 (P<0.001) were found in AML cases. The calculated specificity of CD117 for the diagnosis of AML was 0.97, which was higher than CD13 (0.78) and CD33 (0.75) but comparable to MPO (0.97). The positive predictive value (PPV) of CD117 for AML was 0.99, with the negative predictive value of 0.35. In conclusion, the majority of Thai adult AML cases expressed c-kit. C-kit is infrequently expressed in ALL and appeared to be specific for AML with high PPV. Future targeting therapy using c-kit as a therapeutic target should benefit the majority of Thai AML patients who had high c-kit expression.
Collapse
Affiliation(s)
- Chirayu U Auewarakul
- Division of Hematology, Department of Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkoknoi, Bangkok, Thailand.
| | | | | | | |
Collapse
|
197
|
Sheu LF, Lee WC, Lee HS, Kao WY, Chen A. Co-expression of c-kit and stem cell factor in primary and metastatic nasopharyngeal carcinomas and nasopharyngeal epithelium. J Pathol 2005; 207:216-23. [PMID: 16021677 DOI: 10.1002/path.1822] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Expression of c-kit has been demonstrated in 33% of adult nasopharyngeal carcinomas (NPCs) and in 88% of paediatric NPCs. Patients with tumours expressing c-kit tend to exhibit better survival, but a paracrine/autocrine function for the stem cell factor (SCF)/c-kit system in nasopharyngeal carcinomas has not been reported. This study evaluated the expression of c-kit and SCF by immunohistochemical staining of nasopharyngeal epithelium (NPE) and of primary and metastatic NPCs. In addition, c-kit and SCF expression were studied in HONE-1 NPC cells by immunoprecipitation and western blotting. Expression of c-kit and SCF was detected in 75% and 57% of NPE, respectively, and there was 48% co-expression. In primary NPCs, 86% expressed c-kit, 69% had SCF expression, and there was 67% co-expression. In metastatic NPCs, 76% expressed c-kit, 72% expressed SCF and there was 68% co-expression. Co-expression of c-kit and SCF with tyrosine autophosphorylation of p145(c - kit) was demonstrated in HONE-1 NPC cells. In addition, the expression level of c-kit and its autophosphorylation status was not obviously influenced by the transient co-expression of Epstein-Barr nuclear antigen 1 (EBNA1) and latent membrane protein 1 (LMP1). Co-expression of c-kit and SCF is therefore commonly found in nasopharyngeal epithelium and NPCs, and in HONE-1 NPC cells with autoactivation possibly independent of the co-expression of EBNA1 and LMP1. All of these findings suggest that autoactivation of SCF/c-kit signalling may be a potent regulator of the nasopharyngeal epithelial barrier and of immune function at the nasopharyngeal mucosa surface, and may contribute to the carcinogenesis and progression of NPC. Further molecular analysis is required to evaluate the possibility of treatment with tyrosine kinase inhibitors in NPC, analogous to the treatment of gastrointestinal stromal tumours with STI571.
Collapse
Affiliation(s)
- Lai-Fa Sheu
- Department of Pathology, National Defence Medical Centre, Taipei, Taiwan, Republic of China.
| | | | | | | | | |
Collapse
|
198
|
Sever M, Jones TD, Roth LM, Karim FWA, Zheng W, Michael H, Hattab EM, Emerson RE, Baldridge LA, Cheng L. Expression of CD117 (c-kit) receptor in dysgerminoma of the ovary: diagnostic and therapeutic implications. Mod Pathol 2005; 18:1411-6. [PMID: 16056250 DOI: 10.1038/modpathol.3800463] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The proto-oncogene c-kit encodes a tyrosine kinase receptor, c-kit (CD117), which has been implicated in the development of a number of human malignancies. While the preferential expression of this protein has been well documented in testicular seminomas, there is little data concerning its expression in dysgerminomas of the ovary. We examined the expression of c-kit in 30 cases of ovarian dysgerminomas using immunohistohemical staining with a polyclonal anti-CD117 antibody. Staining was graded in a semiquantitative manner as follows: negative (no staining), 1+(1-10% staining), 2+(10-29% staining), 3+(30-50% staining), or 4+ (>50% staining). Of the 30 cases examined, 26 (87%) demonstrated immunoreactivity for CD117. In total, 10 (33%) demonstrated 4+ staining; 9 (30%) demonstrated 3+staining; 3 (10%) demonstrated 2+staining; 4 (13%) demonstrated 1+staining; and 4 (13%) demonstrated no staining. In conclusion, CD117 immunoreactivity was detected in 87% of ovarian dysgerminomas, a finding that correlates with previously reported frequencies of CD117 expression in seminomas (78-100%). Thus, antibodies to c-kit may be a useful diagnostic marker for ovarian dysgerminoma. Although the prognosis of patients with dysgerminoma is generally good, this receptor could potentially serve as a target for site-specific immunotherapy as an alternative and/or complement to conventional treatment options.
Collapse
Affiliation(s)
- Michael Sever
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Goemans BF, Zwaan CM, Miller M, Zimmermann M, Harlow A, Meshinchi S, Loonen AH, Hählen K, Reinhardt D, Creutzig U, Kaspers GJL, Heinrich MC. Mutations in KIT and RAS are frequent events in pediatric core-binding factor acute myeloid leukemia. Leukemia 2005; 19:1536-42. [PMID: 16015387 DOI: 10.1038/sj.leu.2403870] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Activating mutations in RAS and receptor tyrosine kinases such as KIT and FLT3 are hypothesized to cooperate with chimeric transcription factors in the pathogenesis of acute myeloid leukemia (AML). To test this hypothesis, we genotyped 150 pediatric AML samples for mutations in KIT (exons 8, 17), NRAS and KRAS (exons 1, 2) and FLT3/ITD. This is the largest cohort of pediatric AML patients reported thus far screened for all four mutations. Of the children with AML, 40% had a mutation in KIT (11.3%), RAS (18%) or FLT3/ITD (11.1%), and 70% of cases of core-binding factor (CBF) leukemia were associated with a mutation of KIT or RAS. Mutations in RAS or FLT3/ITD were frequently found in association with a normal karyotype. Patients with a FLT3/ITD mutation had a significantly worse clinical outcome. However, the presence of a KIT or RAS mutation did not significantly influence clinical outcome. We demonstrate that KIT exon 8 mutations result in constitutive ligand-independent kinase activation that can be inhibited by clinically relevant concentrations of imatinib. Our results demonstrate that abnormalities of signal transduction pathways are frequent in pediatric AML. Future clinical studies are needed to determine whether selective targeting of these abnormalities will improve treatment results.
Collapse
Affiliation(s)
- B F Goemans
- Department of Pediatric Hematology/Oncology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Rubin BP, Antonescu CR, Scott-Browne JP, Comstock ML, Gu Y, Tanas MR, Ware CB, Woodell J. A knock-in mouse model of gastrointestinal stromal tumor harboring kit K641E. Cancer Res 2005; 65:6631-9. [PMID: 16061643 DOI: 10.1158/0008-5472.can-05-0891] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A mouse model of gastrointestinal stromal tumor (GIST) has been developed by a knock-in gene targeting strategy, which introduced a Kit gene K641E mutation, originally identified in sporadic human GISTs and in the germ line of familial GIST syndrome patients. Homozygous and heterozygous Kit K641E mice develop gastrointestinal pathology with complete penetrance and all Kit K641E homozygotes die by age 30 weeks due to gastrointestinal obstruction by hyperplastic interstitial cells of Cajal (ICC) or GISTs. Heterozygous mice have less extensive ICC hyperplasia and smaller GISTs, suggesting a dose-response relationship between oncogenically activated Kit and ICC proliferation. Immunoprecipitation and Western blotting reveal GISTs to contain abundant phosphorylated/activated Kit. In addition to ICC hyperplasia and GISTs, homozygous Kit K641E mice exhibit loss-of-function Kit phenotypes, including white coat color, decreased numbers of dermal mast cells, and sterility, indicating that despite its oncogenic activity the mutant form cannot accomplish many activities of the wild-type gene. Kit K641E reproduces the pathology associated with the familial GIST syndrome and thus is an excellent model to study Kit pathway activation, ICC biology, GIST pathogenesis, and preclinical validations of GIST therapies and mechanisms of drug resistance.
Collapse
Affiliation(s)
- Brian P Rubin
- Department of Pathology, University of Washington Medical Center, Seattle, Washington 98195, USA.
| | | | | | | | | | | | | | | |
Collapse
|